Uploaded by Rick K

Feline Dermatology (VetBooks.ir)

advertisement
VetBooks.ir
Chiara Noli · Silvia Colombo Editors
Feline
Dermatology
VetBooks.ir
Feline Dermatology
VetBooks.ir
Chiara Noli • Silvia Colombo
Editors
Feline Dermatology
VetBooks.ir
Editors
Chiara Noli
Servizi Dermatologici Veterinari
Peveragno
Italy
Silvia Colombo
Servizi Dermatologici Veterinari
Legnano
Italy
ISBN 978-3-030-29835-7 ISBN 978-3-030-29836-4
https://doi.org/10.1007/978-3-030-29836-4
(eBook)
© Springer Nature Switzerland AG 2020
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of
the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation,
broadcasting, reproduction on microfilms or in any other physical way, and transmission or information
storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology
now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
VetBooks.ir
Foreword for Feline Dermatology
In 1980, Danny Scott (James Law Professor Emeritus, Section of Dermatology at
Cornell University, New York, USA) published a monograph in the Journal of the
American Animal Hospital Association, entitled Feline dermatology 1900-1978: A
Monograph. This was the first comprehensive survey of skin diseases in the domestic
cat; there had previously been other small descriptive articles and booklets. This was
the first attempt to review all that was known at the time in veterinary science. Since
1980, cats and their skin conditions have always been part of standard textbooks of
veterinary dermatology and veterinary science; Danny Scott published several more
monographs. In 1999, Merial published a book entitled A Practical Guide to Feline
Dermatology, devoted to cats and compiled by a multinational large author group.
While cats make popular pets, they have always been challenging to examine, to
study, to investigate, and to treat. For example, they don’t readily take to diet trials
or accept long courses of oral medications; furthermore, trying to identify clinically
significant allergens remains something of a dark art. They have always been
remarkably independent creatures, and we never really “own them” as a pet. We
remain enthralled with them in part because of their aloof persona as well as their
engaging personalities. In some respects, the level of understanding of their skin
diseases has always lagged behind other domestic animals, especially the dog (with
the old adage that a cat is not a small dog).
Some 20 years after the publication of the book A Practical Guide to Feline
Dermatology, we have this new book Feline Dermatology. The editors have emulated the guide and assembled a large international community of authors who share
their experience and expertise in studying and caring for cats and their skin diseases.
The book comprises three sections. The first section introduces the structure and
function of the skin – the fundamental building blocks that help students and veterinarians to understand the pathogenesis of skin diseases. It is nice to see a chapter on
coat color genetics – a topic often left for other publications and not included with
clinical dermatology textbooks.
The next section provides a series of chapters that discuss the various clinical
presentations of skin diseases in cats with, for example, an approach to the skin
diseases associated with alopecia and so on. Given that cats can present with different cutaneous presentations to the same underlying aetiology, this section is followed by the third section that covers a large array of skin diseases organized by
aetiology.
v
VetBooks.ir
vi
Foreword for Feline Dermatology
The coverage is comprehensive and so there are chapters that will be useful for
students, veterinarians in general practice, residents in training programs, including
dermatology, for veterinarians working in referral centers, and maybe even some cat
owners. There are many illustrations and clinical images that befit the discipline of
veterinary dermatology, which is so reliant on visual representation to appreciate
and understand clinical lesions and the cutaneous reaction patterns that may be
presented.
The editors are to be congratulated on assembling such an array of chapters and
topics, demonstrating that our knowledge and understanding of feline dermatology
has come a long way since the first monographs. This is the first major book on
feline dermatology in many years and should prove to be a useful reference text for
veterinarians for many years to come. Veterinary clinicians may gain a lot of knowledge from the internet but printed books remain popular with publishers and veterinarians. This is one book you ought to have on your shelf.
Aiden P. Foster
Bristol Veterinary School
University of Bristol
Langford, UK
VetBooks.ir
Preface
The world of veterinary dermatology is growing rapidly year after year, as is true for
our knowledge of all animal diseases. The cat is currently receiving great attention
in veterinary medicine: many feline-specific textbooks have been published in
recent years; we have now feline-specific scientific journals, and “feline specialists”
are more and more numerous.
Being veterinary dermatologists with a particular interest in cats, we felt the need
for a feline dermatology textbook. Our aim was to dedicate the appropriate attention
to the cat’s skin and its diseases, which are often peculiar and totally different from
the counterparts described in dogs. A long time has passed by since two previous
feline dermatology books A Practical Guide to Feline Dermatology by Eric
Guaguère and Pascal Prélaud and Skin Diseases of the Cat by Sue Paterson, which
were both published in 1999. After 20 years, it was time for a new feline dermatology textbook and here it is!
This book will hopefully serve both as an essential practical guide for the busy
practitioner, to quickly and surely tackle cats with dermatological conditions, and a
current and complete reference tool for the feline veterinarian and the veterinary
dermatologist.
The most important feline skin diseases such as dermatophytosis and allergic
diseases are described in dedicated chapters. We decided to select different authors
for the majority of the chapters, in order to provide readers with the best possible
review for each subject, written by experts in their specific fields. Each chapter is
greatly enriched with many beautiful colour pictures, which are indispensable to
properly describe a skin disease.
We are very grateful to Springer Nature and all their team for supporting our
project with enthusiasm. Last but not least, we want to say a huge thanks to all the
authors who contributed to this book.
Dedicated to:
Emma, Ada, Luca and all the cats of our lives.
Peveragno, Italy
Legnano, Italy
Chiara Noli
Silvia Colombo
vii
VetBooks.ir
Contents
Part I Introductory Chapters
tructure and Function of the Skin������������������������������������������������������������������ 3
S
Keith E. Linder
oat Color Genetics������������������������������������������������������������������������������������������ 23
C
Maria Cristina Crosta
pproach to the Feline Patient: General and Dermatological
A
Examination������������������������������������������������������������������������������������������������������� 67
Andrew H. Sparkes and Chiara Noli
Part II Problem Oriented Approach to…:
Alopecia�������������������������������������������������������������������������������������������������������������� 95
Silvia Colombo
Papules, Pustules, Furuncles and Crusts �������������������������������������������������������� 109
Silvia Colombo
laques, Nodules and Eosinophilic Granuloma Complex Lesions���������������� 123
P
Silvia Colombo and Alessandra Fondati
xcoriations, Erosions and Ulcers�������������������������������������������������������������������� 137
E
Silvia Colombo
Scaling ���������������������������������������������������������������������������������������������������������������� 149
Silvia Colombo
Pruritus �������������������������������������������������������������������������������������������������������������� 161
Silvia Colombo
Otitis�������������������������������������������������������������������������������������������������������������������� 175
Tim Nuttall
ix
x
Contents
VetBooks.ir
Part III Feline Skin Diseases by Etiology
Bacterial Diseases���������������������������������������������������������������������������������������������� 213
Linda Jean Vogelnest
Mycobacterial Diseases�������������������������������������������������������������������������������������� 251
Carolyn O’Brien
Dermatophytosis������������������������������������������������������������������������������������������������ 265
Karen A. Moriello
eep Fungal Diseases���������������������������������������������������������������������������������������� 297
D
Julie D. Lemetayer and Jane E. Sykes
Sporothrichosis�������������������������������������������������������������������������������������������������� 329
Hock Siew Han
alassezia ���������������������������������������������������������������������������������������������������������� 345
M
Michelle L. Piccione and Karen A. Moriello
Viral Diseases������������������������������������������������������������������������������������������������������ 359
John S. Munday and Sylvie Wilhelm
Leishmaniosis ���������������������������������������������������������������������������������������������������� 387
Maria Grazia Pennisi
Ectoparasitic Diseases���������������������������������������������������������������������������������������� 405
Federico Leone and Hock Siew Han
lea Biology, Allergy and Control�������������������������������������������������������������������� 437
F
Chiara Noli
eline Atopic Syndrome: Epidemiology and Clinical Presentation�������������� 451
F
Alison Diesel
eline Atopic Syndrome: Diagnosis������������������������������������������������������������������ 465
F
Ralf S. Mueller
Feline Atopic Syndrome: Therapy�������������������������������������������������������������������� 475
Chiara Noli
Mosquito-byte Hypersensitivity������������������������������������������������������������������������ 489
Ken Mason
Autoimmune Diseases���������������������������������������������������������������������������������������� 495
Petra Bizikova
I mmune Mediated Diseases������������������������������������������������������������������������������ 511
Frane Banovic
VetBooks.ir
Contents
xi
ormonal and Metabolic Diseases ������������������������������������������������������������������ 531
H
Vet Dominique Heripret and Hans S. Kooistra
Genetic Diseases ������������������������������������������������������������������������������������������������ 547
Catherine Outerbridge
Psychogenic Diseases ���������������������������������������������������������������������������������������� 567
C. Siracusa and Gary Landsberg
Neoplastic Diseases�������������������������������������������������������������������������������������������� 583
David J. Argyle and Špela Bavčar
Paraneoplastic Syndromes�������������������������������������������������������������������������������� 613
Sonya V. Bettenay
I diopathic Miscellaneous Diseases�������������������������������������������������������������������� 627
Linda Jean Vogelnest and Philippa Ann Ravens
VetBooks.ir
Contributors
David J. Argyle The Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, Midlothian, UK
Frane Banovic University of Georgia, College of Veterinary Medicine, Department
of Small Animal Medicine and Surgery, Athens, GA, USA
Špela Bavčar The Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, Midlothian, UK
Sonya V. Bettenay Tierdermatologie Deisenhofen, Deisenhofen, Germany
Petra Bizikova North Carolina State University, College of Veterinary Medicine,
Raleigh, NC, USA
Silvia Colombo Servizi Dermatologici Veterinari, Legnano, Italy
Maria Cristina Crosta Clinica Veterinaria Gran Sasso, Milan, Italy
Alison Diesel College of Veterinary Medicine and Biomedical Sciences, Texas
A&M University, College Station, TX, USA
Alessandra Fondati Veterinaria Trastevere - Veterinaria Cetego, Roma, RM, Italy
Clinica Veterinaria Colombo, Camaiore, LU, Italy
Vet Dominique Heripret CHV Fregis, Arcueil, France
CHV Pommery, Reims, France
Hans S. Kooistra Department of Clinical Sciences of Companion Animals,
Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
Gary Landsberg CanCog Technologies, Fergus, ON, Canada
Julie D. Lemetayer Veterinary Medical Teaching Hospital, University of
California, Davis, CA, USA
Federico Leone Clinica Veterinaria Adriatica, Senigallia (Ancona), Italy
Keith E. Linder College of Veterinary Medicine, North Carolina State University,
Raleigh, NC, USA
xiii
VetBooks.ir
xiv
Contributors
Ken Mason Specialist Veterinary Dermatologist, Animal Allergy & Dermatology
Service, Slacks Creek, QLD, Australia
Karen A. Moriello School of Veterinary Medicine, University of Wisconsin-­
Madison, Madison, WI, USA
Ralf S. Mueller Centre for Clinical Veterinary Medicine, München, Germany
John S. Munday Massey University, Palmerston North, New Zealand
Chiara Noli Servizi Dermatologici Veterinari, Peveragno, Italy
Tim Nuttall Royal (Dick) School of Veterinary Studies, University of Edinburgh,
Roslin, UK
Carolyn O’Brien Melbourne Cat Vets, Fitzroy, Victoria, Australia
Catherine Outerbridge University of California, Davis, Davis, CA, USA
Maria Grazia Pennisi Dipartimento di Scienze Veterinarie, Università di Messina,
Messina, Italy
Michelle L. Piccione School of Veterinary Medicine, University of Wisconsin-­
Madison, Madison, WI, USA
Philippa Ann Ravens Small Animal Specialist Hospital, North Ryde, NSW,
Australia
Hock Siew Han The Animal Clinic, Singapore
C. Siracusa Department of Clinical Sciences and Advanced Medicine, School of
Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
Andrew H. Sparkes Simply Feline Veterinary Consultancy, Shaftesbury, UK
Jane E. Sykes Veterinary Medical Teaching Hospital, University of California,
Davis, CA, USA
Linda Jean Vogelnest University of Sydney, Sydney, NSW, Australia
Small Animal Specialist Hospital, North Ryde, NSW, Australia
Sylvie Wilhelm Vet Dermatology GmbH, Richterswil, Switzerland
VetBooks.ir
Part I
Introductory Chapters
VetBooks.ir
Structure and Function of the Skin
Keith E. Linder
Abstract
Knowledge of skin anatomy and function is fundamental for understanding the
clinical manifestations and impacts of skin diseases. While true for any organ,
this is especially true for the skin, because clinicians can see, touch, and otherwise interrogate the anatomy of this organ directly. Importantly, skin diseases
result from deleterious agents or processes that disrupt specific anatomic components of the skin, and induce physiological responses that distort it, to create skin
lesions. Recognition of skin lesion significance, and thus diseases, is based upon
identifying alterations in normal skin anatomy, including the particular anatomical components that are targeted. Furthermore, the impacts of skin diseases and
treatment choices are understood through knowledge of normal skin functions
and the consequences of its dysfunction. This chapter reviews basic aspects of
feline skin structure and function, with citations from the literature where available, and draws heavily on the comparative information available for humans
and dogs.
The Skin Organ
The skin is organized into multiple, discrete, thin layers that are stacked to create a
sheetlike organ that covers the entire body [1]. Starting externally, the epidermis is
supported by the dermis and then by the panniculus, which connects via fascia to
the underlying musculature or periosteum, for example, in the extremities (Fig. 1).
Nerves and sensory nerve endings invest all three layers variably, whereas blood
K. E. Linder (*)
College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
e-mail: kelinder@ncsu.edu
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_1
3
K. E. Linder
VetBooks.ir
4
Fig. 1 Dorsal mid back, cat. The skin is organized into sheet-like tissue layers. The very thin
epidermis (E) is on the surface and is supported below by the collagenous dermis (D). The panniculus is deepest and is composed of three parts, in areas of the body where all three are present.
The panniculus adiposus (PA) is composed of lobules of adipose, and its most superficial part, the
superficial adipose tissue, is shown here. The collagenous panniculus fibrosis (PF, superficial fascia) supports the panniculus carnosus (PC), which is composed of striated skeletal muscle. Adnexa
are added into these layers, of which hair follicles (HF) are most visible at this magnification. 4X
magnification. Hematoxylin and eosin
vessels are found only in the dermis and panniculus. The skin adnexa (appendages)
are “little organs” added into these three layers multifocally during development
and include, for example, hair follicles, skin glands, and claws. All three skin layers
are highly modified to create discrete anatomical structures like the planum nasale
and footpads.
The skin thickness, made of the dermis and epidermis together, varies by body
region and is only generally 0.4–2.0 millimeters thick in the cat, being thicker on
the dorsal body and proximal limbs and thinner on the ventral body, distal limbs,
and ears [2]. These layers are the thickest on the footpads and planum nasale [2].
The panniculus varies greatly in thickness, being absent to >2 centimeters, depending on the degree of adiposity of the patient and the anatomical region of the body;
it is generally thickest on the ventrum, especially in obese patients, thinner on the
dorsum, and progressively thins to become mostly absent on the extremities.
Structure and Function of the Skin
5
VetBooks.ir
Epidermis
The epidermis is remarkably thin (Fig. 2) and measures only 10–25 micrometers in
truncal areas, but is thicker on footpads (Fig. 3) and planum nasale [2, 3]. In most
body areas, the viable epidermis contains only three to five keratinocyte layers.
The superficial nonviable epidermis, the stratum corneum, contains more numerous
cell layers composed of very thin cells, called corneocytes, which are less than 1
micrometer thick (Fig. 2). Haired areas tend to have thinner epidermis than non-­
haired areas.
The epidermis is a stratified cornifying epithelium composed of keratinocytes
(85%) arranged in four layers based on morphology: the stratum basale, stratum spinosum, stratum granulosum, and stratum corneum (Fig. 2) [1]. Keratinocytes continually proliferate in the basal epidermal layer, then migrate, and differentiate to
form the upper epidermal layers and finally shed (desquamate) from the skin surface.
The epidermis also contains resident Langerhans cells, migrating T-lymphocytes,
and uncommon neuroendocrine Merkel cells (<1%) [1]. Melanocytes are present
in pigmented epidermis and are absent in areas of white spotting. In the cat, mast
cells are rare in the epidermis but can move into the epidermis in greater numbers
during inflammatory diseases such as allergic skin diseases. Nerves extend into the
epidermis but blood vessels do not.
Fig. 2 Face, cat. The epidermis is composed of four morphological layers: stratum basale (SB),
stratum spinosum (SS), stratum granulosum (SG), and stratum corneum (SC). The deep stratum
corneum, called the stratum compactum (arrowheads), is very thin and formed by compact
orthokeratosis. The superficial stratum corneum, called the stratum dysjunctum (arrows), is
expanded mostly by histology artifact into a basket weave pattern of orthokeratosis. The basement
membrane zone (BMZ; location of the ultramicroscopic basement membrane) connects the
epidermis to the dermis (D). Fibrocytes (F) and mast cells (MC) are in the dermis. 100X
magnification. Hematoxylin and eosin
K. E. Linder
VetBooks.ir
6
Fig. 3 Carpal footpad, cat. Footpads (including digital pads) have a thick epidermis, with a robust
stratum corneum (SC), and thick dermis (D). Hair follicles (HF) and sebaceous glands are absent
in footpads but are present in haired skin at the footpad margin (left of the image). Footpad
cushions (C) are discrete modifications of the panniculus and contain small lobules of adipose with
robust fibrous septa. Eccrine glands (EG) are embedded in the cushion, and eccrine ducts (ED) exit
directly through the dermis and epidermis to empty onto the footpad surface. 4X magnification.
Hematoxylin and eosin
The deepest epidermal layer, the stratum basale (stratum germinativum), contains epidermal stem cells with mitotic activity and continually supplies new keratinocytes to all epidermal layers (Fig. 2) [1, 4]. Basal layer keratinocytes are smaller
and more cuboidal, with less cytoplasm, and attach the epidermis to its basement
membrane and thus to the dermis. Moving up, the stratum spinosum is named for
the spinous projections observed on keratinocyte membranes with paraffin section
histology – an artifact of tissue processing that accentuates desmosomal attachments between cells. Spinous layer cells are larger due to more abundant cytoplasm,
are polyhedral, and have more visible cytoplasmic keratin intermediate filaments.
Next, the stratum granulosum is named for cytoplasmic, basophilic keratohyalin
granules that are visible with hematoxylin and eosin (H&E) staining and store
mostly proteins, like profilaggrin, needed for cornification [4]. Lamellar bodies,
not visible with paraffin histology, form in this layer as well and deliver lipids,
enzymes, and other key components to the extracellular surface during cornification [4]. The stratum corneum, the external most layer of the epidermis, forms by
terminal differentiation called cornification, which creates nonviable corneocytes
from the viable granular layer keratinocytes below [4]. During this process, keratinocytes loose most of their cytoplasmic water and organelles and flatten to become
very thin (less than 1 micrometer) discoid cells with linear faceted (5–6) margins.
The nucleus is also lost, and thus the cornification is orthokeratotic. On paraffin
histology, the deep corneocytes are densely compacted into a discrete layer called
the stratum compactum, and the superficial corneocytes are separated on their faces,
VetBooks.ir
Structure and Function of the Skin
7
because of processing artifact, in an open basket weave pattern in a layer called the
stratum dysjunctum (Fig. 2) [5]. Corneocytes are continually shed from the body in
a process called desquamation.
In the stratum corneum, corneocytes are stacked into many layers, approximately
10 to 15 on the trunk and 50+ on the footpads and planum nasale, and are sealed by
intercellular lipids [3]. Fewer corneocyte layers are present in haired, low friction
areas and more are in high friction areas such as palmar and plantar footpad surfaces. On the trunk, corneocytes are stacked into uniform vertical columns, overlapping only slightly at their margins, whereas on the footpads, corneocyte stacking is
nonuniform and cells overlap extensively and variably, creating greater cell-to-cell
surface contact, which is thought to increase adhesion. Intercellular lipids, delivered by lamellar bodies, are highly organized into a stack of lipids called the lipid
envelop, which seals the entire extracellular space and creates the most important
barrier preventing external water loss from the skin.4 These lipids are composed of
ceramides, cholesterol, and fatty acids. Certain lipids, like linoleic acid, are essential and are very important for lipid envelop formation and function. Corneocytes
are continually shed from the skin surface by desquamation. Desquamation occurs
because the normal physiochemical environment (pH, hydration, etc.) of outer
stratum corneum promotes activation of numerous intercellular enzymes to cleave
corneodesmosomes and degrade intercellular lipids, which allows corneocytes to
separate away [4].
Clinically, a buildup of the stratum corneum on the skin surface, either due to
increased production of corneocytes or altered desquamation, is called scaling.
Partial loss of the epidermis leads to an erosion, which causes water loss from the
skin surface. Eroded epidermis appears smooth and slightly moist due to the missing stratum corneum, which is responsible for normal epidermal surface architecture and barrier function. Eroded epidermis lacks hemorrhage as the epidermis does
not contain blood vessels. In contrast, complete loss of the epidermis and the basement membrane is an ulcer, which appears moist to wet and granular (because of
collagen exposure and recruited leukocytes and fibrin), and it often contains hemorrhage because of exposed dermal blood vessels.
Epidermal Basement Membrane
The epidermal basement membrane (basal lamina) is composed of numerous
filamentous proteins and proteoglycans that bind together to form an ultrathin,
mesh-like sheet that supports the basal cells and blankets the dermis [6]. Basal
cells are structurally connected by hemidesmosomes to the basement membrane,
which in turn is connected to the dermis by anchoring fibrils composed of collagen
VII. Basement membrane zone is used to refer to this structure on light microscopic
histology because it is too thin to be directly visualized (Fig. 2).
Epidermal strength results from physical interconnections between cytoskeletal
proteins, cellular adhesion complexes (desmosomes and hemidesmosomes), and the
VetBooks.ir
8
K. E. Linder
epidermal basement membrane [6]. The cytoskeleton of each keratinocyte is linked
by desmosomes, and in basal keratinocytes, it is linked to the basement membrane
by hemidesmosomes. The keratinocyte cytoskeleton contains large amounts of keratin intermediate filaments, which are bundled together like ropes to form tonofilaments with high tensile strength. The buildup of specialized keratin filaments in
each epidermal layer is called keratinization, a key part of cellular differentiation
in the epidermis. Desmosomes of the stratum granulosum are modified by addition of corneodesmosin, and by other changes, to become corneodesmosomes in
the stratum corneum [4]. Many diseases of epidermal fragility, i.e., mechanobullous diseases and pustular diseases, cause skin lesions by disrupting desmosomes,
hemidesmosomes, or the basement membrane.
Dermis
The dermis (corium) is a thick, discrete, organized layer of extracellular matrix
(collagens, etc.) that provides structure, toughness, and flexibility to the skin,
and it supports the epidermis and adnexa as well as blood vessels, lymphatic
vessels, and nerves found within it (Fig. 2) [1]. The dermis is divided into a
thin superficial papillary layer with more loosely arranged matrix and finer collagen bundles and a thicker deep reticular layer that is more densely packed
with coarser collagen bundles. The dermis is composed primarily of collagen,
mostly types I and III, for strength, elastin for elasticity, and proteoglycans,
like hyaluronic acid, for hydration and turgor pressure. In cats, the dermis has a
scalloped deep margin (Fig. 1) with projections that connect to the lobular septa
of the panniculus below. Dermal vessels are arranged in three sheet-like plexuses of arteries and veins that are located just below the epidermis, in the mid
dermis, and in the deep dermis at the junction with the panniculus [7]. The dermis contains microscopic bundles of smooth muscle attached to hair follicles,
called erector pili muscles, and free bundles in the dermis of teats (nipples) and
scrotum [1, 2]. Also in the scrotum, the dartos tunic of the testis extends to the
panniculus where it contributes smooth muscle and collagenous stroma. Small
bundles of skeletal muscle extend to the dermis in facial and perineal areas
only. Adipocytes are not normal constitutes of cat dermis and are part of the
panniculus.
Mesenchymal cells maintain the dermal matrix and include fibrocytes (Fig. 2),
which are spread individually throughout the dermis, as well as pericytes and
Schwann cells that are localized around blood vessels and nerves, respectively. Low
numbers of immune cells such as mast cells, dermal dendritic cells, lymphocytes,
and basophils can be found in a healthy dermis where they are usually individualized and localized more to superficial perivascular and less to interstitial areas. Mast
cells are common in the dermis of cats with 4 to 20 mast cells visible per 400x
microscopic field on histology (Fig. 2) [8]. Neither neutrophils nor eosinophils are
found in normal dermis or epidermis.
Structure and Function of the Skin
9
VetBooks.ir
Panniculus
The panniculus (hypodermis, subcutis) is composed of discrete sheetlike layers
of adipose, muscle, and fascia (Fig. 1) [1, 2, 9]. Immediately below the dermis,
the panniculus adiposus (called the superficial adipose tissue) contains adipose
arranged into lobules by thin fibrous septa (Fig. 1) [9]. Deeper, the panniculus fibrosus (superficial fascia) is a thin, variably discrete, sheet of fibrous tissue that connects to the lobular septa of the panniculus adiposus. Coursing within the fascia is
a thin layer of striated muscle called the panniculus carnosus (cutaneous trunci) [1,
2]. The panniculus carnosus is more developed dorsally on the trunk (Fig. 1), neck,
and proximal limbs and tapers away on the ventral abdomen (Fig. 4) and limbs to
become absent on the extremities. Depending on the body region, like the extremities, the panniculus fibrosus merges with the deep fascia that surrounds the muscle
of the skeleton or periosteum [8]. However, in some areas, like the ventral trunk,
another layer of lobular adipose (called the deep adipose tissue) is present below the
Fig. 4 Ventral mid abdomen, cat. The panniculus has three main layers: panniculus adiposus, panniculus carnosus, and panniculus fibrosus. The panniculus adiposus is composed of lobules of
adipose just below the dermis, called the superficial adipose tissue (SAT), and in some body
regions, a second layer which is deeper, called the deep adipose tissue (DAT), is also present. The
panniculus fibrosus (PF) is a sheet of fibrous tissue (superficial fascia) that connects to the thin
fibrous septa of adipose and supports the panniculus carnosus (PC), which is diminished ventrally.
40X magnification. Hematoxylin and eosin
VetBooks.ir
10
K. E. Linder
panniculus fibrosus that is an additional deeper portion of the panniculus adiposus
(Fig. 4) [9]. The panniculus adiposus is the thickest on the trunk, especially on the
ventrum of the cat, where it can be measured in centimeters in obese patients, and
it is mostly absent in the extremities. The panniculus is specialized to form cushions in footpads (Fig. 3), which are composed of lobules of adipose with thickened
fibrous septa.1.2 Arteries, veins, nerves, and lymphatic vessels are present in the panniculus and pass through to the dermis above.
Skin Adnexa (Skin Appendages)
Hair Follicles
Hair follicles produce hairs that cover nearly the entire body of the cat except
for small areas like the mucocutaneous junctions, external genitalia, teats, planum nasale, and footpads [1, 2]. The density of hairs on the cat is higher, 25,000
per square centimeter, compared to the dog, 9000 per square centimeter, but the
density varies by breed and anatomical location. Most hair follicles in the cat are
a compound type in which several hair follicles share a single follicle opening (follicular ostium), while fewer are of a simple type with one hair follicle per ostium.
Primary hair follicles are larger and produce larger hair shafts (guard hairs, outer
coat hairs), while secondary hair follicles are smaller and produce smaller hair
shafts (undercoat hairs). In cats, most hair follicles are grouped such that a single,
simple, large primary hair follicle (central primary hair) is surrounded by two to
five compound follicles, each with a primary hair(s) (lateral primary hair(s)) and
3–12 secondary hairs, with numbers partly depending on age [1, 2, 10, 11]. The
tail lacks this arrangement and hair follicles are larger [2]. Primary hairs of cats are
much thinner, 40–80 micrometer in diameter, compared to dogs, 80–140 micrometer, whereas the secondary hairs of cats are 10–20 micrometer and those of the
dog are 20–70 micrometer. Most hair follicles lay in the skin at an angle to the epidermal surface such that their hair shafts all point caudally on the head and trunk
and distally on the limbs (Fig. 1). In the dermis, the ectal side (outside) of the hair
follicle is closer to the epidermis (acute angle), and the ental side (inside) is further
from the epidermis (obtuse angle). Specialized sinus hair follicles (vibrissae or
whiskers) are very large simple hair follicles surrounded by a blood sinus and have
complex innervation and sensory tactile function (slow-adapting mechanoreceptor) (Fig. 5) [1, 2, 8]. Sinus follicles produce vibrissae that are large tactile hairs
that vary greatly in length. Sinus hairs are found on the face (muzzle, eyebrows,
lips), and palmar carpus, and variably on the neck, forelimbs, and paws of cats
and are arranged individually, in small clusters, or in short rows in some areas on
the face. A second tactile hair type, the tylotrich hair, arise from follicles that are
slightly larger and more richly innervated than those of primary hair follicles and
contact an adjacent sensory tylotrich pad (touch dome) when compressed [1, 2].
Tylotrich hairs are scattered individually, in low density, throughout most of the
haired skin.
VetBooks.ir
Structure and Function of the Skin
11
Fig. 5 Sinus hair follicle
(vibrissae follicle), face,
cat. The sinus follicle is a
very large simple hair
follicle that produces a
large hair shaft (HS), or
whisker, and is named for a
large blood-filled sinus (S)
that surrounds the follicle.
The sebaceous gland (SG)
and dermal papilla (DP)
are annotated. 4X
magnification.
Hematoxylin and eosin
Hair follicles form during development as specialized epithelial down-growths
of the epidermis (ectodermal origin) interacting with clusters of specialized mesenchymal cells (mesodermal origin) called the dermal papilla [10]. A fully formed
hair follicle is a linear, layered, tubular epithelial structure that opens superficially
at the follicular ostium and forms a solid bulb at its deep base with an invagination
that encircles the dermal papilla (during anagen only) [1]. The erector pili muscle
is a smooth muscle, and it originates in the dermis from the epidermal basement
membrane and inserts on the ental side of the hair follicle [1]. This muscle elevates
the hair shaft on the skin surface, for example, in behavior responses and with cold
temperatures to trap more insulating air in the hair coat. The hair follicle epithelium
is encased in a basement membrane (the glassy membrane) that is surrounded by
a thin layer of collagen and specialized dermal fibrocytes, called the dermal root
sheath or fibrous sheath [1]. The perifollicular dermis is richly supplied by small
blood vessels that branch from all three dermal plexi but most prominently the mid
dermal plexus [7]. Hair follicles in the anagen phase can extend into the panniculus
adiposus (Fig. 1).
Hair follicles continually cycle to produce, hold, and shed hairs [10, 12]. The hair
growing phase (anagen, Fig. 6) transitions through a short involution phase (catagen) and then ends in a resting phase (telogen, Fig. 6) in which a hair is retained or
kenogen in which a hair is not retained (also called hairless telogen) [12]. A resting hair shaft is actively shed (exogen) usually when the cycle begins again. Hair
K. E. Linder
VetBooks.ir
12
Fig. 6 Rostral chin, cat. Large primary hair follicles in the growing anagen phase and the resting
telogen phase of the hair follicle cycle. A) In fully developed anagen, the hair bulb (HB) encases
the dermal papilla (DP) and actively produces the hair shaft (HS) and inner root sheath (IRS). B)
In fully developed telogen, the hair bulb and internal root sheath are absent and the external root
sheath (ERS) regresses to surround the hair shaft, while the dermal papilla (DP), remains connected by only an epithelial strand (ES). The hair shaft stops growing, and its pointed end (club
hair) is sealed by brightly eosinophilic trichilemmal cornification. 20X magnification. Hematoxylin
and eosin
shedding in the cat is mosaic (nonsynchronous) [10]. The duration of phases varies
by age, breed, season, etc. [13] For example, the length of the hair shaft depends on
the length of the anagen phase – longer hair is due to a longer anagen phase.
The hair follicle has three zones (segments) called the infundibulum, isthmus, and
inferior portions [12]. The infundibulum is the superficial, permanent, non-­cycling
segment that morphologically resembles the epidermis and attaches to it [1]. The
isthmus and the deeper inferior portions change morphologically with the hair follicle cycle and have five main components, some only being present during the anagen
phase (Fig. 6) [1, 12]. First, the inner root sheath surrounds the central follicle lumen
and has its own three layers, an inner cuticle, Huxley’s layer, and outer Henle’s layer.
The raised exposed edges of overlapping cuticle cells point internally (toward the
hair bulb) and interlock with opposite facing hair shaft cuticle cells. The inner root
sheath is only present during anagen (Fig. 6) when its keratinocytes continually
migrate up in concert with the growing hair shaft, cornify, and shed to the infundibular lumen. Second, the companion layer is a single layer of cells that separates the
VetBooks.ir
Structure and Function of the Skin
13
inner rooth sheath from the external root sheath. Third, the external root sheath is
several keratinocytes thick, it encases the inner root sheath, and it is contiguous with
the infundibulum. Forth, the hair bulb, forms during anagen and is composed of hair
matrix cells arranged in concentric layers that generate each of the layers of the inner
root sheath, the companion layer, and the hair shaft (Fig. 6). Finally, the fourth part,
the derma papilla (follicular papilla), is encased by an invagination of the hair bulb
(Fig. 6). The dermal papilla is composed of mesenchymal spindle cells, blood vessels, and nerves, and its molecular communication with the hair matrix cells partly
controls follicle cycling, hair shaft formation, and hair shaft pigmentation.
The hair shaft is formed by cornification of hair bulb cells (hair matrix cells), which
makes it rigid, and contains three concentric layers, the outer cuticle, the cortex, and
the inner medulla [1]. The hair shaft cuticle is a single layer of overlapping flattened
cells in which exposed cell edges point outward (away from the hair bulb). The cortex
is compacted and is nonpigmented or variably pigmented. The medullary cells have
an open structure that in some follicles highlights an empty nuclear profile – primary
hairs have a medulla but secondary hairs do not. The medulla may be pigmented or
non pigmented. The outer end of the hair shaft is pointed in a long thin taper, while
the inner end (hair root) is either connected to the soft viable hair bulb during anagen
or is sealed-­off in telogen by trichilemmal cornification to form a short, rigid, pointed
taper with a rough surface (club hair) (Fig. 6).
Clinically, hair shafts are epilated and examined microscopically (trichogram) to
identify the stage of the hair follicle cycle, primary or secondary status, and any hair
shaft abnormalities. Anagen phase hair bulbs indicate active hair growth and, on
trichogram, are recognized to be soft, flexible, rounded, and often axially deviated
and pigmented when hair is pigmented. Telogen phase hairs (club hair) indicate resting hair follicles and have short tapered ends that are rough externally, rigid, and not
axially deviated and are nonpigmented in hair that is pigmented or nonpigmented.
Skin Glands
In the cat, sebaceous glands are small, simple or compound, lobulated alveolar glands
that connect to the lower infundibular lumen of hair follicles (pilosebaceous unit) by
a very short duct lined by stratifed and cornifying epithelium [1, 2, 14]. At the edge
of lobules (peripheral zone), a single thin layer of cuboidal reserve cells divides and
differentiates to form larger polygonal lipid-vacuolated cells called sebocytes centrally (maturational zone), which shed to the lumen (holocrine secretion) to form
sebum. In the cat, cytoplasmic vacuoles of sebocytes are very small and very uniform in size. Larger, often multilobulated, sebaceous glands are present on the face,
especially the chin (Fig. 7; submental organ), ear base, dorsum, anal-rectal junction,
palmar carpus (carpal gland), and interdigital paw skin. Meibomian glands (tarsal
glands) are large sebaceous glands of the eyelid margin, especially the upper eyelid
(Fig. 8) [2]. Sebaceous glands are not found in the planum nasale or the footpads.
Apocrine glands (epitrichial sweat glands) and eccrine glands (atrichial sweat
glands) in cats are simple coiled tubular glands that secrete via a duct to the deep
VetBooks.ir
14
K. E. Linder
Fig. 7 Rostral chin, cat.
Sebaceous glands (SG) in the
chin area (submental organ)
are very large and
multilobulated, and the
dermis (D) is expanded to
support the larger sebaceous
glands in addition to the
apocrine glands (AG), hair
follicles (HF), and epidermis
(E). 4X magnification.
Hematoxylin and eosin
Fig. 8 Upper eyelid, cat. The
upper eyelid is lined by
haired skin (HS) externally
and by mucosa (M) of the
palpebral conjunctiva
internally. Meibomian glands
(MG) are enlarged sebaceous
glands that align in a single
long row that tracks the
mucocutaneous junction. 4X
magnification. Hematoxylin
and eosin
infundibulum of primary hair follicles (epitrichial) (Fig. 7) and to the footpad
surface (atrichial) (Fig. 3) [1, 2, 14]. Glands are lined by cuboidal to low columnar cells that secrete by the release of apical blebs of the cytoplasm to the gland
lumen and then to a thin duct lined by a bilayer of short cuboidal cells. A few
myoepithelial cells surround the gland. Ceruminous glands are modified apocrine
glands in the external ear canal (see below). Eccrine glands are not found on the
planum nasale.
VetBooks.ir
Structure and Function of the Skin
15
Fig. 9 Proximal dorsal
tail, cat. The dorsal tail
gland (DTG) of the cat,
also called the supracaudal
gland, is non-discrete and
is composed of hepatoid
glands on hair follicles
along most of the dorsal
tail. Erector pili muscles
(M) are largest on the
proximal dorsal tail and
originate from the
basement membrane of the
epidermis (E) and insert on
hair follicles in the dermis
(D). 4X magnification.
Hematoxylin and eosin
In the cat, the dorsal tail gland (Fig. 9, supracaudal gland) is formed by hepatoid
glands located on hair follicles of the dorsal tail, especially proximally [15]. Feline
hepatoid glands are a mixed lipid and protein secretion type and thus appear very
pale, eosinophilic, and moderately vacuolated in hematoxylin and eosin-stained
histology sections compared to brightly eosinophilic and non-vacuolated hepatoid
glands (circumanal glands) of the dog, which produce primarily protein [15]. This
is also the situation for hepatoid glands present on the anal sacs of cats.
Anal sacs (perianal sinuses) are paired in the cat and are located in the subdermal tissue of the perineum bilaterally (Fig. 10) [1, 2]. The anal sac and its short,
duct-­like, narrow opening to the anal-rectal skin junction are thin walled and lined
by stratified squamous epithelium with an orthokeratotic pattern of cornification,
which are supported by a thin layer of dermal matrix. Apocrine glands and large
hepatoid glands of the anal sac (Fig. 10) are grouped in this matrix along the anal
sac margin and empty to it [2, 14].
The mammary gland is a compound tubulo-alveolar gland arranged by septa into
lobules and lobes where each gland empties via a branched ductular system to a teat
(nipple) [1]. Glandular secretions pass through intralobular, to interlobular ducts,
to lactiferous ducts, and then to a teat sinus (teat cistern), all of which are lined by
either a simple layer or a bilayer of cuboidal cells. In the cat, the teat sinus empties
externally through four to seven papillary ducts that are lined by stratified squamous
epithelium. The teat dermis contains free bundles of smooth muscle but scant other
adnexa. In the cat, four mammary glands are organized in linear mammary chains
on the right and left side of the ventral abdomen.
VetBooks.ir
16
K. E. Linder
Fig. 10 Anal sac and
associated glands, cat. The
anal sac (AS) is thin walled
and lined by stratified
squamous epithelium.
Multifocal apocrine glands
(AG) and hepatoid glands
(HG) empty to the anal
sac. 4X magnification.
Hematoxylin and eosin
Claws
The cat claw is a very specialized and complex structure composed of a cornified
claw sheath (claw horn, claw) that is formed by stratified epithelium and supported
by specialized dermis (corium) [16]. The claw in the cat is a highly tapered (sharply
pointed) curved, and ventrally flattened cone that has a rounded wall dorsally, flattened blades on its slides, and a narrow cutting ridge ventrally. Proximally, a band
of claw matrix cells continually divide and differentiate to supply keratinocytes
for growth of the claw. More distally, the claw bed (claw plate) cells provide sliding adhesion that allows epithelial cells to move distally and to cornify into the
rigid claw. Cats sharpen their claw tips by repeat shedding of a cornified horn cap
that is promoted by scratching [16] – shed horn caps are sometimes mistaken for a
sloughed claw. The claw encases the claw dermis and the closely apposed unguicular process of the third phalanx. The hard cornification of the rigid claw is bordered
by soft cornification where it is contiguous with the skin fold (claw fold). The claw
fold is large on the dorsal and lateral margins of the claw and minimal below. Below
the claw centrally, a small sole first merges with the narrow skin fold, which merges
with palmar or plantar digital pad [16]. The claw fold is modified and elaborated
(claw sac, etc.) in the cat to allow claw retraction.
External Ear (Chapter, Otitis)
The skin of the pinna and external ear canal (external acoustic meatus) are lined
by stratified squamous epithelium with an orthokeratotic pattern of cornification,
which is supported by a thin dermis [2, 17]. Skin adnexa are present on all surfaces
but are smaller and less densely placed in the inner ear pinna (concave pinna) and
especially in the external ear canal in comparison to the outer pinna (convex pinna)
(Figs. 11 and 12). Hair follicles and sebaceous glands are in all of these locations
17
VetBooks.ir
Structure and Function of the Skin
Fig. 11 External pinna, cat. The external pinnae contain a sheet of elastic cartilage (EC) centrally
that is lined by dermis (D) and epidermis (E) on the convex (top of image) and concave (bottom of
image) sides. Adnexa, including hair follicles (HF), sebaceous glands, and apocrine glands, are
more numerous and larger on the convex surface compared to the concave surface. 4X magnification.
Hematoxylin and eosin
Fig. 12 Horizontal ear canal
(cross section), cat. The ear
canal is lined by a thin
epidermis (E) and dermis.
The inset demonstrates the
small sparse adnexa of the ear
canal in the dermis (D),
including a hair follicle (HF),
a sebaceous gland (SG), and
a ceruminous gland (CG). 4X
magnification and 20x
magnification (inset).
Hematoxylin and eosin
but are in a low density in the external ear canal. Apocrine glands are present on the
convex and concave pinna. Modified apocrine glands, called ceruminous glands,
are in the portion of the external ear canal that is supported by the annular cartilage
(Fig. 12) and are more numerous in the deep one third of the canal [17–20]. These
glands connect to the sparse follicles or directly to the epidermis [17]. Ceruminous
gland secretions mix with sebum, epidermal surface lipids, and desquamated corneocytes to form a waxy protective material called cerumen. Centrifugal epithelial
migration of keratinocytes off of the external tympanic membrane and on to the
external ear canal helps to clear cerumen from the surface of the tympanic membrane, which is located at the deep extent of the ear canal [19].
18
K. E. Linder
VetBooks.ir
Skin Pigmentation
The skin derives its color from pigmentation (melanin pigment); blood in vessels
(red heme pigment); the endogenous reflective properties of the epidermis, dermis,
and adnexa; as well as the quality of light being reflected [8]. Melanin pigment is
composed of two types, brown to black eumelanin and red to yellow pheomelanin,
that are expressed variably to produce a color range. From neural crest, melanocytes migrate during development into the epidermis, follicles, and claws and then
produce melanin pigment in membrane-bound cytoplasmic organelles called melanosomes. Through dendritic processes, a melanocyte transfers pigmented melanosomes to a certain number of local keratinocytes, and together, they are called an
epidermal melanin unit (or follicular melanin unit in the hair bulb). The amount
and type of melanin produced and the degree of dispersal of melanosomes to keratinocytes affect the pigment intensity, ranging from a pale dilute to a very dark
color. Melanocytes in the anagen hair bulb deliver melanosomes to growing hair
shafts, continuously or episodically, and the latter produces color banding (agouti,
etc.), which is partly controlled by the dermal papilla. Many coat color variations
(Chapter, Coat Color Genetics) in cats are due to inheritance of alleles that alter
melanocyte distribution or presence/absence, melanosome dispersal, and/or the
amount and type of melanin produced. Clinically, the loss of pigment (leukoderma,
leukotrichia) is due to disruption of the epidermal melanin unit, and/or follicular
melanin unit, and thus can result from diseases that injure melanocytes and/or
keratinocytes.
General Skin Functions
The skin has many important functions that, when compromised by disease, have
significant consequences for the patient [8].
Physical Barrier Function
The skin protects the body from physiochemical injury. It prevents entry of foreign
materials, parasites, and infectious agents while, at the same time, preventing loss
of water and fluid components (electrolytes, macromolecules, etc.) from the body.
To do this, the epidermis and dermis provide the toughness of the skin, and hair
reduces frictional injury. The panniculus provides a cushion to injury especially
in footpads. Skin pigment and hair block damaging solar radiation. The stratum
corneum, especially the lipid envelop, seals the epidermis to water loss, whereas
deeper epidermal layers also contribute, for example, via tight junctions in the stratum granulosum. Corneocytes are shed continuously from the skin surface, eliminating attached microorganisms. Claws even serve as offensive physical defense
against attack of other animals and as tools needed by cats for climbing and handling of prey.
Structure and Function of the Skin
19
VetBooks.ir
Immune Defense
More than just a passive physical barrier, the skin immune system actively identifies, blocks, and eliminates pathogens through actions of the innate (keratinocytes,
mast cells, basophils, natural killer cells, dendritic cells, sebum, etc.) and acquired
immune skin systems (T-cells, B-cells, dendritic cells, etc.). Additional cells, neutrophils, eosinophils, and macrophages are recruited to the skin through its vasculature
and contribute to skin defense and immune function. Sebum and stratum corneum
constituents contribute to skin surface pH, and fatty acid composition favors skin
colonization by beneficial bacterial commensals and limits pathogens. Interestingly,
the skin helps to maintain peripheral immune tolerance, by assisting the thymus in
educating the acquired immune system on self and non-self antigens.
Thermoregulation
The skin is a key organ of thermoregulation that both works to prevent heat loss
and to promote it, as needed, to optimize core body temperature. The hair coat and
adipose are the main thermal insulating barriers, and the former can be modified
by erector pili muscles moving the hair and controlling its density. Skin blood flow
is actively promoted, restricted, and/or shunted to alter core heat transfer to the
skin, especially in the distal limbs and ears. Pigment in hair and epidermis absorbs
light energy, leading to heating of the skin. Sweating promotes cooling through
evaporation.
Metabolic Functions
The skin has numerous metabolic functions; many maintain skin homeostasis, while
others also serve systemic functions. For example, vitamin D is activated in the epidermis via exposure to sunlight. And, after further activation in the liver and kidney,
vitamin D impacts epidermal proliferation and differentiation in the skin as well as
contributes to calcium homeostasis of blood and bone, among many other functions,
systemically. Expression of p450 enzymes in the epidermis means that xenobiotic
compounds can be processed there. The panniculus adiposus contributes much of
the bodies’ capacity to store energy in the form of lipids. Similarly, dermal collagen
is a protein reservoir. The epidermis, hair follicles, and skin glands produce useful
substances but also eliminate endogenous and exogenous metabolic constituents,
such as some toxins (lead in hair).
Communication
The skin glands produce scents that are important for olfactory communication in
carnivores. Erector pili muscles, especially along the back and tail, elevate the hair,
VetBooks.ir
20
K. E. Linder
changing the physical hair coat appearance, to visually communicate behavior status and warning signals to other animals, and to disperse pheromones. Skin and hair
coat pigmentation, although modified dramatically in many domestic cats by human
selection pressures, provide camouflage important for carnivores while hunting.
Sensory Perception
The skin is a major organ of sensation, and its sensory nerve endings distinguish
temperature (hot and cold), pain, pruritus, burning, touch, etc.
References
1. Monteiro-Riviere N. Integument. In: Eurell JA, Frappier BL, editors. Dellmann’s textbook of
veterinary histology. 6th ed. Iowa: Blackwell Publishing Professional; 2006. p. 320–49.
2. Strickland JH, Calhoun ML. The integumentary system of the cat. Am J Vet Res.
1963;24:1018–29.
3. Monteiro-Riviere NA, Bristol DG, Manning TO, et al. Interspecies and interregional analysis
of the comparative histologic thickness and laser Doppler blood flow measurements at five
cutaneous sites in nine species. J Invest Dermatol. 1990;95:582–6.
4. Matsui T, Amagai M. Dissecting the formation, structure and barrier function of the stratum
corneum. Int Immunol. 2015;27:269–80.
5. Bowser PA, White RJ. Isolation, barrier properties and lipid analysis of stratum compactum, a
discrete region of the stratum corneum. Br J Dermatol. 1985;112:1–14.
6. Hammers CM, Stanley JR. Mechanisms of disease: pemphigus and bullous pemphigoid. Annu
Rev Pathol. 2016;11:175–97.
7. Meyer W, Godynicki S, Tsukise A. Lectin histochemistry of the endothelium of blood vessels
in the mammalian integument, with remarks on the endothelial glycocalyx and blood vessel
system nomenclature. Ann Anat. 2008;190:264–76.
8. Miller WH, Griffin CE, Campbell K. Muller & Kirk’s small animal dermatology. 7th ed. St.
Louis: Elsevier; 2013. p. 1–56.
9. Stecco C. Subcutaneous tissue and superficial fascia. In: Functional atlas of the human fascia.
Philadelphia: Elsevier; 2015. p. 21–30.
10. Meyer W. Hair follicles in domesticated mammals with comparison to laboratory animals
and humans. In: Mecklenburg L, Linek M, Tobin D, editors. Hair loss disorders in domestic
animals. Iowa: Wiley-Blackwell; 2009. p. 43–61.
11. Zanna G, Auriemma E, Arrighi S, et al. Dermoscopic evaluation of skin in health cats. Vet
Dermatol. 2015;26:14–7.
12. Welle MM, Wiener DJ. The hair follicle: a comparative review of canine hair follicle anatomy
and physiology. Toxicol Pathol. 2016;44:564–74.
13. Ryder Ryder ML. Seasonal changes in the coat of the cat. Res Vet Sci. 1976;21:280–3.
14. Jenkinson DM. Sweat and sebaceous glands and their function in domestic animals. In: von
Tscharner C, Halliwell REW, editors. Advances in veterinary dermatology, vol. 1. Philadelphia:
Bailliere Tindall; 1990. p. 229.
15. Shabadash SA, Zelikina TI. Detection of hepatoid glands and distinctive features of the hepatoid acinus. Biol Bull. 2002;29:559–67.
16. Homberger DG, Ham K, Ogunbakin T, et al. The structure of the cornified claw sheath in the
domesticated cat (Felis catus): implications for the claw-shedding mechanism and the evolution of cornified digital end organs. J Anat. 2009;214:620–43.
VetBooks.ir
Structure and Function of the Skin
21
17. Strickland JH, Calhoun ML. The microscopic anatomy of the external ear of Felis domesticus.
Am J Vet Res. 1960;21:845–50.
18. Fernando SDA. Microscopic anatomy and histochemistry of glands in the external auditory
meatus of the cat (Felis domesticus). Am J Vet Res. 1965;26:1157–61.
19. Njaa BL, Cole LK, Tabacca N. Practical otic anatomy and physiology of the dog and cat. Vet
Clin North Am Small Anim Pract. 2012;42:1109–26.
20. Tobias K. Anatomy of the canine and feline ear. In: Gotthelf L, editor. Small animal ear diseases, an illustrated guide. 2nd ed. St. Louis: Elsevier-Saunders; 2005. p. 1–21.
VetBooks.ir
Coat Color Genetics
Maria Cristina Crosta
Abstract
The various breeds of cats differ considerably one from the other, not only in
terms of their different morphological features but also in terms of the colour,
length, structure and texture of their coat. The feline coat has various functions,
such as aesthetic and mimetic, heat regulation, perception of the body position
through vibrissae and tylotrich pads, social and sexual communication, and acts
as a barrier against mechanical, physical and chemical insults. In the first part of
this chapter, the morphology and cycle of the hair are briefly introduced, including melanin synthesis. In the second part, the genetics of hair length, structure,
texture, colour and colour patterns are detailed, providing a good description to
understand the specific functional aspects of the feline coat.
The various breeds of cats differ considerably one from the other, not only in terms
of their different morphological features but also in terms of the colour, length,
structure and texture of their coat.
The Coat
Cat show organisers classify cat breeds according to three main categories:
• Longhair cats, the representatives being the Persian (in all colour shades and
varieties), the British Longhair, the Selkirk Rex and the Highland Fold
Images by Lia Stein
M. C. Crosta (*)
Clinica Veterinaria Gran Sasso, Milan, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_2
23
VetBooks.ir
24
M. C. Crosta
• Medium-length hair cats, e.g. Norwegian Forest, Maine Coon, Balinese, Birman
• Shorthair cats, e.g. European, Chartreux, Russian Blue, British Shorthair
Within these categories, the coat is then classified according to pattern, colour
and colour distribution.
Function
The coat has various functions:
• Aesthetic and mimetic;
• Heat regulation, in function of the length, thickness and density of the coat, as
well as of its colour and shine (a light-coloured coat reflects light better and
allows to keep body temperature constant);
• Perception of body position (vibrissae, tylotrich pads);
• Social and sexual communication, thanks to both the visual effect and as support
to pheromones;
• Barrier against mechanical, physical and chemical insults.
Whatever its length, a cat’s coat is made to protect the animal and help it adapt
to its environment, like in the case of cats that live in very cold climates. The coat of
these cats (Maine Coon, Norwegian Forest cats) consists of long primary hairs
(basic coat) and a thick undercoat.
The hair of a Norwegian Forest cat is water-repellent. This trait makes its coat
especially suited for the adverse weather conditions of its country of origin. In
shows, sometimes judges assess this feature by dripping some water over its coat.
Another example is the Turkish Van, a cat that has a very thick coat in winter and
that sheds in a spectacular manner in summer. In fact, in this season it loses almost
all of its fur, to the point of looking like a shorthair cat. This breed has adapted to
the climate of Central Anatolia, its region of origin, where there is a great difference
in temperature between winter (−20 °C) and summer (+40 °C).
Morphology
Macroscopically speaking, regardless of length, cat hair can be classified as:
1. Primary hair (guard hair)
2. Secondary hair (down hair).
Like all carnivores, cats have compound hair follicles. This means that the coat
is comprised of many small units. Each unit consists of two to five larger hairs
VetBooks.ir
Coat Color Genetics
25
(primary hairs) surrounded by clusters of smaller hairs (secondary hairs). To each
primary hair are associated from five to twenty secondary hairs.
Each primary hair has its own sebaceous gland, a sweat gland and an arrector pili
muscle. A primary hair emerges through the surface of the skin through its own
independent infundibulum. Secondary hairs are associated only to one sebaceous
gland and emerge from a common infundibulum.
It is estimated that, in cats, for each square cm of skin there are anywhere from
800 to 1600 of these units.
From a functional point of view, hair can also be classified as:
1. Protection hair: these hairs are straight and thicker.
2. Intermediate hair: these hairs are thinner than protection hairs and more variable
in cross diameter. They grow in the opposite direction of the underhair and play
an isolating and protective role together with the underhair.
3. Underhair: these hairs are short and thin and have a crimped appearance, sometimes curly. In winter, they trap warm air and create a veritable insulating barrier
against the cold, while in summer they limit the absorption of heat coming from
the outside.
The proportions vary according to breed:
• All three hair types can be present, but they can be highly modified (e.g. Devon
Rex).
• One can be missing (e.g. protection hair in the Cornish).
• One can be more abundant than the others (e.g. underhair in Persians).
• One can be very scarce (e.g. underhair in the Korat).
There are two types of tactile hairs:
• Vibrissae: they grow on the muzzle, around the eyes, in the throat region and on
the palmar face of the carpi. These hairs are thick and contain specialised nerve
structures.
• Tylotrich hairs: they are distributed all over the body and consist of a larger than
normal hair follicle containing a single short hair and surrounded by a capsule of
neurovascular tissue at the level of the sebaceous gland. These hairs are believed
to be slow-adapting mechanoreceptors.
The hair shaft is made up of three concentric structures, the medulla, the cortex
and the cuticle.
The medulla is the inner structure of the hair and consists of longitudinal rows of
cells that are solid close to the root and that progressively fill with air and glycogen
as they rise towards the tip.
The cortex forms the middle structure of the hair and is formed by hard and fuse-­
shaped cells with the longer axis parallel to the hair’s axis. These cells contain the
pigment that gives the hair its colour.
VetBooks.ir
26
M. C. Crosta
The cuticle is the outermost structure of the hair and is made up of scales (in
humans they are imbricate, like the tiles of a roof, while in cats they are triangular
with a spinous edge and with the hooked free edge facing the direction of the hair
tip).
Hair Growth Cycle
The hair follicle is the structure from which the hair grows. It consists of an
upper section called ‘infundibulum’, an intermediate section called ‘isthmus’,
and a deep section that is the ‘bulb’. The infundibulum and the isthmus are the
permanent portions of the hair, while the bulb is present only in the active growth
phase.
The hair bulb consists of matrix cells (that generate the hair itself and the
internal sheath that contains the root) and of pigment-producing cells called
melanocytes.
The hair follicle has an active, cyclic growth phase called anagen and a rest phase
called telogen. They are separated by a transition phase called catagen.
The duration of the anagen phase is hereditary and determines the final length of
the hair. In this phase, the dermal papilla is very well developed and the cells of the
bulb’s matrix actively multiply to form the hair. The bulb’s melanocytes actively
produce pigment (melanin) and distribute it to the hair’s cells that progressively
migrate towards the surface of the skin.
During the transition phase called catagen, pigment production stops completely and the production of cells by the matrix progressively slows down to a
stop. The last cells produced therefore are entirely pigment-less and this explains
why in this phase of the cycle the section of the hair closest to the skin is also the
lightest.
During telogen, the follicle, in a resting phase, has shrunk to one third of its
length and the dermal papilla has transformed into a small mass of undifferentiated
cells. Hair shedding does not occur simultaneously throughout the coat but according to a ‘mosaic’ shedding pattern. This is because neighbouring hair follicles are
all at different stages of growth. The hair grows to a pre-set length that can vary
according to the body region and is genetically determined.
The active growth phase and therefore hair growth speed is highest in summer
and lowest in winter. Indeed, it is thought that in summer 50% of the follicles are in
telogen, while in winter this percentage rises to 90%.
Melanin Synthesis
Melanin is the pigment responsible for colouring skin and hair. This is not its only
function, however. By distributing through the cytoplasm, it protects the cells of
VetBooks.ir
Coat Color Genetics
27
Fig. 1 The synthesis of
eumelanin and
pheomelanin
L-Tyrosine
Tyrosinase
+Cu
L-DOPA
Tyrosinase
+Cu
DOPAQUINONE
+ Cysteine
Pheomelanin
Eumelanin
the epidermis and of the deeper skin layers from ionizing radiation and from ultraviolet (UV) light. Melanin also eliminates the toxic free radicals generated by the
skin’s cells following exposure to sunlight and in the course of inflammatory
processes.
Melanin synthesis is genetically determined (Fig. 1). Its production may be
stimulated by several factors, such as exposure to ultraviolet radiation from the
sun, and may be affected by hormone unbalance. There are various types of
melanin, but the basic types are eumelanin and pheomelanin. Eumelanin granules are contained in melanosomes and are responsible for the brown-blackishblack colour. Pheomelanin granules are also contained in melanosomes and
impart a yellow-brownish-red colour. There are many intermediate variations
between these two types. Pheomelanin features a higher sulphur content compared to eumelanin. Although different, eumelanin and pheomelanin have a
common metabolic life. An enzyme called tyrosinase in the presence of oligoelements, such as copper, transforms tyrosine first into DOPA and then into dopaquinone and from there, through a sequence of oxidations, into the various kinds
of melanin pigment. The importance of the role played by this enzyme in
VetBooks.ir
28
M. C. Crosta
pigment synthesis is proven by the fact that the mutation of the structural gene
of tyrosinase is responsible for many forms of albinism in humans and in animals. The synthesis of eumelanin requires high concentrations of tyrosinase,
while that of pheomelanin needs lower concentrations but requires cysteine.
Tyrosinase is heat-sensitive, which means that its concentration decreases in
proportion to the increase in temperature. In body areas where the temperature
is lower (legs, for example) thanks to the increase in enzyme activity there is a
higher deposit of eumelanin granules, and this gives the coat a darker coloration. This explains why, when the coat of a black cat is observed closely in the
trunk area, a lighter coloration is seen at the base, while in the muzzle areas and
on the legs (that are colder areas), the colour is darker.
The Genes Controlling Hair Length, Structure and Texture
Although from a morphological viewpoint we distinguish three kinds of cat coats –
long, semi-long and short – from a genetic viewpoint, in terms of hair length, the coats
are two – shorthair and longhair (Figs. 2, 3 and 4). The genes described in this chapter
are summarized in Table 1. A definition of genetic terms is provided in Box 1.
Fig. 2 Long-haired cat
Coat Color Genetics
VetBooks.ir
Fig. 3 Semilong-haired
cat
Fig. 4 Short-haired cat
29
30
M. C. Crosta
VetBooks.ir
Table 1 Main genes controlling coat colour in cats
Genes controlling distribution of coat colour
A Agouti
a non-agouti
Original wild colour, hair with alternated
The colour bands on the single hairs disappear,
light and dark bands
and the result appears solid colour
Genes controlling colour
B Black
b brown (or chocolate)
bl light brown (or cinnamon)
o non-orange
O Orange – sex-linked because it is located on the
X chromosome
w normal colour
W White dominant epistasis
Genes controlling colour intensity
C full Colour – even intensity all over
cb burmese
body
cs siamese
ca albino blue eyes
c albino pink eyes
Genes controlling colour density
D Dense – normal hair colour density
d dilution or maltesing
Genes controlling hair colour development
i complete development of pigment in
I Inhibition of development of pigment in hair
hair
wb (or ch) no tipping
Wb (or Ch) tipping
Genes controlling white spot distribution
s normal colour distribution in coat – no
S piebald white Spotting – more or less extensive
white spots
white spots
G normal colour distribution in coat – no
g ‘gloves’ of the birman
gloves
Genes controlling tiger tabby patterns
T mackerel Tabby (wild type)
Ta abyssinian tabby
tb blotched tabby (or classic tabby)
Genes controlling and modifying hair length, structure and texture
L
shorthair
l
longhair
R normal coat
r
Cornish Rex
Re normal coat
re Devon Rex
Ro normal coat
ro Oregon Rex
rd normal coat
Rd Dutch Rex
rs normal coat
Rs Selkirk Rex
Hr normal coat
hr Sphynx, Bambino, Elf, Dwelf
hrbd normal coat
Hrbd Don Sphynx, Peterbald, Levkoi
wh normal coat
Wh Wirehair
VetBooks.ir
Coat Color Genetics
Box 1
• Homozygote/Heterozygote
A homozygote is an individual having two identical alleles for one same
trait (one coming from the mother and the other from the father). In fact, for
each single trait each individual receives a pair of ‘corresponding’ genes
called alleles, one inherited from each parent. If the individual inherits two
identical alleles, that individual is homozygous for that specific trait (e.g. BB
or bb). Conversely, if the individual inherits two different alleles from its parents, the individual will be a heterozygote (Bb).
• Dominant and Recessive
When an allele manages to express itself (phenotypic expression) both in a
homozygous individual (BB) and in a heterozygous individual (Bb or Bbl), it
is said to be dominant. If an allele expresses itself only in the homozygote, it
is called recessive and is indicated with the lower case letter (bb).
The dominant trait does not allow the recessive trait (b) to express itself.
Two cats, one BB and the other Bb, shall both have a black coat, but BB is
homozygous black while Bb is a black carrier of the chocolate colour that,
being recessive to B, cannot be expressed. This is why in a genotype, 2 letters
are used to indicate a trait (e.g. BB, Bb). If instead only one letter is used followed by a dash (e.g. B-), it means that we don’t know whether the individual
is homozygous (BB) or heterozygous (Bb) for that same trait. In genetics, the
dominant allele is indicated with an upper case letter, and usually uses to be
the first letter of the gene to which it refers (B for Black, D for Dense).
• Polygenes
These are a group of genes (also called ‘modifiers’) the single action of
which is often not quantifiable, but when working together have a cumulative
effect and can change the action of the main gene. They affect quantitative
traits (size, hair length, etc.), often quite significantly.
• Epistasis
Some genes have the capacity to prevent other genes from expressing themselves. Pheomelanin, for example, masks eumelanin; the non-agouti gene covers
the tabby patterns; the W gene (dominant white) masks the expression of all of
the other genes responsible for coloration and colour distribution (Fig. 31).
31
VetBooks.ir
32
M. C. Crosta
Sometimes, the epistatic effect of the W gene is not totally effective. One often
sees white kittens with a spot of colour (black, blue cream, etc.) on their head.
This spot of colour, that totally disappears by the age of ten months or so, is nothing but the hidden colour the kitten will pass on to its progeny as an adult. All
cats, independent of colour, are genetically tabby, i.e. they possess striping in
their genotype (Fig. 32). In ‘self’ (solid colour) cats, striping is present but not
visible because the a gene (non-­agouti) does not allow striping to express itself
(epistatic effect). The O gene (orange) transforms colour pigments into pheomelanin and through epistasis deactivates the loci coding for the production of
eumelanin (Fig. 33). Moreover, the a gene (non-agouti) makes striping disappear
(epistasis) only in eumelanic coats and has no effect on pheomelanic coats. This
is why, in red cats, striping is always present. It is difficult to obtain an evenly
red-coloured coat with intense red coloration (breeders have a hard task in doing
this) because quite often residual stripes show up on the muzzle, tail and legs.
Sometimes, in the attempt to diminish the undesired striping, varieties of red
coats with excessively bleached coloration are selected. Unlike shorthair cats, in
which the stripes are more evident, in the Persian these ‘defects’ are corrected by
the hair length hair. Just like the other solid colour coats, the red coat must be
even, i.e. each single hair must have the same intensity from the root to the tip,
and its coloration must be as red as possible. Next to solid colour coats, also
known as self, breeders select red tabbies in which the stripes are actually highlighted, in a curious game of contrast between the red background of the coat and
the intense and prominent red of the pattern.
• Density and Dilution
Colour density is given by the dominant gene D responsible for dense pigmentation. Pigment granules are deposited one by one and evenly along the
cortex and medulla of the hair. The entire surface of the granule reflects light,
giving the hair a darker coloration. Coat coloration dilution is given by the
recessive d gene (or Maltese gene) that causes a different distribution in space
of the pigment granules without changing their shape. This different layout
causes less light refraction and therefore the colour appears lighter.
• Incomplete Dominance
Incomplete dominance occurs when, in a pair of alleles, one allele does not
totally dominate the other and the resulting individual shows intermediate
traits (e.g. Tonkinese).
• Agouti and Non-agouti
This is an Indian term used to indicate a South American rodent. In genetics, it is used to describe the wild coloration of some mammals. The Agouti
VetBooks.ir
Coat Color Genetics
33
gene codes for the multiple banding of each single hair with yellow-greyish
bands and a darker hair tip (ticking). Agouti allows the tabby alleles to express
themselves. Agouti is the background colour against which one sees the
stripes of a tabby coat (Fig. 34). The non-agouti gene codes for the masking
of the bands of yellow-greyish colour on each single hair, making it still
banded, but dark and very dark. Therefore, the coat appears to the eye of a
single colour. It has an epistatic effect on tabby alleles.
• Eumelanin, Pheomelanin and Tyrosinase
The eumelanin granules determine brown, blackish or black coloration (B,
bb, blbl). The pheomelanin granules determine red, yellow or orange coloration (O-). Eumelanin and pheomelanin are formed starting from the amino
acid tyrosine. This process occurs thanks to the action of the enzyme tyrosinase (heat-sensitive) that oxidates tyrosine into various intermediate compounds (DOPA, dopaquinone). The C gene (colour intensity) codes for the
enzyme’s correct structure, which means that its inactivation by high temperature is much slower than its production and, therefore, melanin is regularly
produced and can provide full coloration of the entire coat. The Albino alleles
(cb and cs) progressively cause a structural change in tyrosinase, making it
especially heat-sensitive. In the warmer areas of the body, due to a lower
influence of tyrosinase, there will be less pigment deposit (the body is warmer
and therefore the coat colour will be paler), while in the cooler areas of the
body (the extremities), there will be a greater deposit of pigment and therefore
darker coloration. In the Burmese (cb), the enzyme’s structural modification
causes the coat to change from black to dark brown and the eyes to become
yellow or amber. In the Siamese coat, the cs gene makes the tyrosinase even
more sensitive to heat, and therefore the difference between the colour of the
body and that of the points (extremities) is much more evident and the eyes
become blue (Fig. 35). The ca and c genes, instead, cause the destruction or
lack of production of the enzyme, and therefore the coat is totally white and
the eyes are blue and pink, respectively.
Length Genes
• short hair: L (dominant)
• long hair: l (recessive)
The original coat is short and is governed by a dominant gene labelled L. Long
hair is given by its recessive mutant allele and is labelled l. Gene l is responsible not
only for the long hair in Persian cats but also for the semi-long coats of the Maine
Coon cat, Norwegian Forest cat, Siberian cat and Burmese cat. The various coat
VetBooks.ir
34
M. C. Crosta
lengths are due to the presence of polygenes or modifier genes. These are minor
genes the single effect of which is too small to be observed. However, when they are
acting together with other genes, they produce observable effects because together
they are variably capable of changing the action of the main gene. Hair length is not
the only trait to be taken into consideration. Structure and texture, too, are important
elements of evaluation when examining the various kinds of coats. The coat can be
more or less thick and abundant, and the three types of hair (protection hair, intermediate hair and underhair) can be normal and all present at the same time.
Sometimes, like in the Devon Rex, there are significant changes, or one of the three
types of hair can be missing, such as the protection hair in the Cornish Rex. The
types of hair can be present in different proportions, like in the Korat that has almost
no underhair or in the Persian that abounds in underhair, compared to the amounts
of protection and intermediate hair.
Structure and Texture
•
•
•
•
•
•
•
•
r (recessive) Cornish Rex/German Rex
re (recessive) Devon Rex
ro (recessive) Oregon Rex
Rd (dominant) Dutch Rex
Rs (dominant) Selkirk Rex
Wh (dominant) American Wirehair
hr (recessive) Sphynx/Bambino/Elf/Dwelf
Hrbd (dominant) Don Sphynx/Peterbald/Levkoy
The most significant changes regarding hair structure and texture involve the r,
h, Wh and Hrbd genes.
r genes
Cornish Rex
A cat famous for its coat is the Cornish Rex. Its peculiar coat is due to the presence
of the r gene. This gene codes for the lack of protection hairs and for deep changes
in intermediate hair and underhair. This cat’s fur is very soft, dense, coarse to the
touch, wavy and so curly that the coat looks like that of a sheep’s fleece. Even its
facial and supraorbital vibrissae are curled.
Devon Rex
This cat’s coat is due to a recessive gene called re (Fig. 5). All three hair types are
present but deeply modified. The coat is less wavy and curly than that of the Cornish
and in general its coat has a more sparse appearance. The facial and supraorbital
vibrissae may be broken or even absent. The r and re genes are recessive mutant
Coat Color Genetics
35
VetBooks.ir
Fig. 5 Devon Rex cat
genes located on different loci of the chromosome, and by crossing a Cornish Rex
with a Devon Rex one can obtain non-rex coated kittens.
There are other modifications regarding the r genes, especially those of the
Oregon Rex, a cat with a coat deriving from the presence of the ro gene, a recessive mutant allele that causes the protection hair to disappear. The coat of the
German Rex is caused by the presence of the r gene, identical to that of the
Cornish. In the Dutch Rex and in the Selkirk Rex, instead, the genes are two
dominant mutant alleles: Rd and Rs, respectively. The Dutch Rex is currently not
being bred. The Selkirk rex has a thick, plush and curly coat which can be short
or long.
36
M. C. Crosta
VetBooks.ir
h genes
Sphynx/Bambino/Elf/Dwelf
The Sphynx is a cat that has no protection or intermediate hairs due to the presence
of the recessive hr gene. The sparse underhair, that can even be totally absent, is
located on the muzzle, on the outside base of the ears, on the feet, scrotum and tail.
The skin is very soft to the touch, resembling suede, and wrinkles are present on the
face, between the pinnae and on the shoulders. The cat is medium size, muscular,
with wide chest and rounded abdomen. The cross-breeding of the Sphynx with
other breeds has resulted in the Bambino, Elf and Dwelf breeds. The Bambino cat
is the result of the cross between the Sphynx and the Munchkin (‘Sausage cat’), a
short-legged cat. The Bambino is a smaller version of the Sphynx has no hair, a long
chest and a rounded abdomen. The hind legs are longer than the front legs, the
muzzle is triangular with wide and tall pinnae. The cross between the Sphynx and
the American Curl has generated the Elf breed, a naked, tall and muscular cat with
prominent cheek bones, like the Sphynx cat. This cat also presents curled pinnae,
like the American Curl breed. The breeding of the Elf with the Munchkin/Bambino
has given rise to the Dwelf breed (the name is a blend of ‘dwarf’ and ‘elf’, the
mythical creature with pointed ears). This cat is small, naked, short-legged and has
curled pinnae like the Elf cat.
Hrbd genes
Donskoy/Peterbald/Levkoy
This group includes the Donskoy (Don Sphynx), the Peterbald and the Levkoy. The
gene responsible for these breeds is Hrbd, a dominant gene located on a locus different from that of the hr gene. The Donskoy is a middle-sized cat with cuneiform
head, wide ears with rounded tips positioned high on the head and medium to long
legs. The Donskoy cat is preferable naked; however it occasionally may have hairs
called ‘flock’ when less than 2 millimetres long and ‘brush’ when more than 2 millimetres long. The hair is sparse and hard all over the body, with naked areas on the
head, upper neck or on the back. These cats, with residual hair, cannot participate in
cat shows but are successfully employed for reproduction. The Peterbald derives
from the cross of the Don Sphynx with the Siamese/Oriental Shorthair. It has all the
morphologic features of the Siamese/Oriental Shorthair (long, elegant and slender
body with long legs) but carries all the cutaneous features of the Donskoy, with
wrinkles on the face, between the ears and on the shoulder. As for the Donskoy, the
naked cats are preferred, but ‘haired’ cats can be used for reproduction. The Levkoy
comes from the cross between the Donskoy and the Scottish Fold. The Levkoy may
be naked or have residual hair and its pinnae are folded forward like the ones of the
Scottish Fold. Again, the naked ones are preferred, but kittens and younger cats can
sometimes present some residual hair. Crossing between these breeds and the
Sphynx is not allowed.
Coat Color Genetics
37
VetBooks.ir
Wh Genes
Wirehair
This group includes a cat breed that has a very particular coat, the American
Wirehair, which owes its coat to the dominant mutant gene Wh. All three hair types
are present but they appear modified and curly, which makes the Wirehair’s coat
hard and coarse to the touch.
The Genes Controlling Coat Coloration and Patterns
Tabby
The transmission of cat coat colours follows precise genetic rules according to Mendel’s
laws. If when speaking of hair length the shorthair is the original coat, when speaking
of colour patterns all cat coats derive from the tabby. The tabby coat is the most commonly found in nature because it is highly mimetic. Indeed, tabby is the original wild
coat, the primordial coloration from which all other coat colours have descended by
mutation. The name ‘tabby’ derives from ‘Attabi’, the characteristic Baghdad district
famous for making the precious striped silk cloth called ‘taffetà’. This name, later
shortened to ‘tabby’, was then used to describe the striped coat of cats. In tabby cats,
the stripes seem drawn against the coat’s background that is commonly called Agouti.
Agouti and Non-agouti
Agouti is an Indian word used to indicate a rodent that lives in the rain forests of
Central and South America, subsequently used in genetics to describe the wild coloration of the hare and rabbit. Agouti (A) in the genotype determines a ‘striped’ or
‘banded’ coloration of the coat via a pigment synthesis system called on-off. Darker
colour bands produced during the on phase alternate with lighter colour bands produced during the off phase. In this way, each single hair is not of a single colour but
is marked by alternated dark and light colour bands and has a dark-­coloured tip
(Fig. 6). Its recessive mutant allele, non-agouti, represented by the symbol a, suppresses the light-color banding, which is substituted by a dark colored band different
from the first one. The hair appears to the eye as self-coloured (solid) because the
bands cannnot be distinguished. The difference between a tabby coat and a “self”
(solid) coat is well represented by the coat of the leopard and the black panther. As
many will know, the leopard and the black panther are the same animal. However, in
the leopard the black patches are very visible on the yellow coat, while in the black
panther the patches are black on a black background and therefore cannot be appreciated. The non-agouti gene manages to make the light colour bands disappear only
with eumelanic colours, but has no effect on pheomelanic colours (in practice, a red
non-­agouti cat has a striped coat). Agouti allows the striping coat to appear, which
means that what one sees in a striped coat is a complex coloration deriving from two
colour components and controlled by two different gene groups: Agouti + Tabby.
38
M. C. Crosta
VetBooks.ir
Fig. 6 Agouti hair coat
Gene A not only affects coat coloration but also that of the skin and nose. In eumelanic cats, the nose leather is not one solid colour, like in solid coloured cats, but
rather brick red/pink/old rose coloured, rimmed with the coat’s basic colour.
Patterns
Agouti is the basic colour, i.e. the background on which the pattern seems to be
drawn. There are four main patterns associated with the three tabby genes:
––
––
––
––
Ticked tabby or Abyssinian
Spotted tabby
Mackerel tabby
Blotched tabby
The tabby genes are T, Ta and tb, and they are autosomic (three different alleles
on the same locus). They are capable of generating striped coat patterns that are
called markings:
––
––
––
––
Ta is responsible for the ticked tabby coat (or Abyssinian coat).
T is responsible for the mackerel tabby and for the spotted tabby coat.
tbtb is responsible for the Blotched tabby or classic tabby coat.
Ta is considered dominant over T, T is dominant over tb, and tb is recessive to
both. These coats in heterozygous specimens (such as Ttb or TaT) are not as
well defined and precise as homozygous coats (TaTa or TT).
T is responsible for both the mackerel and the spotted tabby patterns. There are
many theories to explain this. Some claim that the spotted coat is derived from a
polygenic action; others state it derives from the presence of other genes capable of
breaking up and rounding the stripes of mackerel coats.
icked Tabby or Abyssinian (Ta Gene)
T
In this coat, the Agouti is distributed all over the coat and for this reason all of the
coat appears evenly ‘ticked’. Each hair has regularly alternating bands of different
VetBooks.ir
Coat Color Genetics
39
colour (Fig. 7). The root is apricot colour while the tip is of the so-called basic
colour, that can be black, chocolate, blue, cinnamon or fawn. The more bands on
the hair, the more appreciated the coat. This colour is most frequent in the cats of
the savannah and in wild cats living in arid and desert areas, while in selected
breeds it is typical of the Abyssinian, of the Singapura and of the Ceylon cat. In
some breeds, the presence of stripes on legs, neck, muzzle and tail are considered
defects, as in the Abyssinian, while in others they are indispensable, as in the
Singapura.
ackerel Tabby (T Gene)
M
Mackerel is the term used to indicate the tabby coat with uninterrupted vertical lines
(Fig. 8). ‘Mackerel’ is a fish that has thin parallel stripes that descend from its back to
its midline. The cat’s coat has a straight and uninterrupted black line along the spine,
Fig. 7 Abyssinian cat
Fig. 8 Mackerel tabby cat
VetBooks.ir
40
M. C. Crosta
running from the back of the head to the base of the tail. On its sides, shoulders and
thighs, there are distinct narrow, continuous and parallel stripes. Its legs, tail and neck
are well banded. These cats have an ‘M’ on their forehead and two or three lines that
follow the profile of the cheeks. They have many small spots on their underside, from
the throat to the belly. The ‘M’ on the forehead has inspired quite a few legends. One
tells of the baby Jesus trembling in the manger despite being wrapped in blankets.
Mary called in all the animals to warm Him but Jesus still trembled. Then a tabby cat
appeared and snuggled up to Him in the manger, covered Him with his body and
warmed Him. As a sign of her gratitude, Mary drew an M on its forehead. Another
legend states that a snake crawled into the sleeve of the Prophet Mohammed’s robe,
and a tabby cat killed it immediately. From that moment onwards, all tabby cats were
born with the M on their forehead to remind everyone that these cats deserve respect.
potted Tabby (T Gene)
S
The cat with a spotted coat has many small round or oval spots on its coat, separated
one from the other and evenly distributed (Fig. 9). A thin, straight and uninterrupted
Fig. 9 Spotted tabby cat
VetBooks.ir
Coat Color Genetics
41
black line may be present from the back of the head to the base of the tail. An M is
designed on the forehead and two or three lines follow the profile of the cheeks. The
neck shows two uninterrupted stripes, while the legs and tail are banded. They have
many small dots on their abdomen, from the neck to the belly. This coat is also called
‘maculate’ and is typical of the Egyptian Mau and of the Ocicat. The ‘resetting’ on
the coat of the Bengal can be considered a modified spotted trait.
lotched Tabby (tb Gene)
B
Also known as classic tabby. This is the showiest and most spectacular of coats
because the Agouti background is marked by a butterfly-shaped pattern, the upper
and lower wings of which are clearly designed on the flanks and shoulders of the
cat (Fig. 10). Along the spine, from the back of the head to the base of the tail, there
are three large stripes, a central one flanked by another two, distinctly separated
and parallel to the first. The forehead bears an M and two or three lines follow the
profile of the cheeks. The neck is decorated by two uninterrupted stripes while the
legs and tail are banded. They have many small spots on their abdomen, from the
neck to the belly. The coat of the Marbled Bengal is considered a classic tabby
modified.
ifferences Between Tabby and Self-Coloured
D
• Nose: in self-coloured cats, the colour of the nose is solid. In tabby cats, the nose
colour is brick red, pink, old rose, and rimmed with the coat’s basic colour.
• Chin: in tabby cats, the colour of the chin is lighter than in self cats.
Fig. 10 Blotched tabby
cat
VetBooks.ir
42
M. C. Crosta
• Eyes: the eyes of tabby cats are rimmed with the coat’s basic colour (and sometimes the lips are too) and a halo of lighter colour immediately around the eye.
• Ears: in self-coloured cats, the whole ear is evenly coloured, while in the tabby,
and especially in tabby Points, a ‘thumbmark’ (or pouce) is present, i.e. a lighter
area of colour on the external base of the ear.
Self or Solid Colours
The original colour is the tabby coat due to the association of the Agouti gene with
the tabby alleles. All that was needed was the mutation of the Agouti gene (A) into
non-agouti (a) to make the grey-yellowish bands on every single hair to disappear
and generate the solid colour coat. Non-tabby cats, usually called self or solid, are
cats that have coat of a single colour with hair that is evenly coloured from the root
to the tip. The colour of each single hair is defined by the colour genes. Gene B is
the colour-coding gene that allows melanosomes to produce eumelanin granules
that give the hair a dark coloration. Gene B produces the hair’s black
pigmentation.
Gene B has two recessive mutant alleles called b (brown or chocolate) and bl
(light brown or cinnamon). In these mutations, the pigment granules become
deformed until they take on oval (b) or even more elongated (bl) shapes. The granules deformed in this way reflect the light, giving the hair a lighter colour: b produces the colour chocolate while bl the colour cinnamon. Black, B, is the dominant
shape, while b and bl are both recessive to gene B, and bl is recessive to b
(B > b > bl).
Dilution
These colours exist in the dilute form. In fact, thanks to the action of the dilution
gene also known as the Maltese gene (d), the pigment granules inside the cortex
aggregate and take on a different distribution. By doing this, they reflect the
light and the coat appears of a lighter colour, allowing black to become blue,
chocolate to become lilac and cinnamon to become fawn. Solid coat colours
thus are six:
Non-diluted
Black
Chocolate
Cinnamon
Diluted
Blue
Lilac
Fawn
Black
The hair should be black from the root up and not contain any traces of brown or
have any white hairs or grey underhair. Often the hair tips when exposed to the sun,
VetBooks.ir
Coat Color Genetics
43
or the collar that is easily soiled by food and water, tend to become reddish or brown
coloured. Nose leather and paw pads are black (Fig. 11).
Blue
The coat that ranges from a very light grey colour to slate grey is called blue. The
most desirable is the lighter colour, as even as possible, from the tip to the root,
without black tips or white hairs. Nose leather and paw pads should be blue. It is a
dilution of black (Fig. 11).
Chocolate
The coat is milk chocolate coloured with a warm and even hue from the tip to the
root of the hairs, without stripes or hairs of other colours. The nose leather is milk
chocolate coloured, while the paw pads range from milk chocolate to cinnamon
rose. Chocolate is a mutation of black (Fig. 12).
Lilac
Also called lavender or frost, lilac is a dilution of chocolate. The coat appears evenly
pinkish-light grey without any kind of striping (Fig. 13). Nose leather and paw pads
are coloured pinkish lavender.
Cinnamon
The brown coat is very light: this is a mutation of black (Fig. 14). In the Abyssinian
and in the Somali, this colour is called sorrel.
awn
F
It is a dilution of cinnamon (Fig. 15).
Red and Tortoiseshell
Gene O transforms colour pigments into pheomelanin and deactivates the eumelanin production loci. Pheomelanins are granules that produce a red/orange colour.
The orange gene is located on the X chromosome and for this reason is defined
‘sex-linked’. Pheomelanic colours do not express all of the changes in hues seen
Fig. 11 Black and blue
kittens
44
VetBooks.ir
Fig. 12 Chocolate cat
Fig. 13 Lilac cat
M. C. Crosta
Coat Color Genetics
45
VetBooks.ir
Fig. 14 Cinnamon cat
Fig. 15 Fawn cat
with eumelanic colours, but only the colours red and cream. The cream colour is
obtained through the intervention of dilution genes.
There is a very special coat colour known as ‘tortoiseshell’. In this coat, the
colours red and black are perfectly mixed or are present as clearly separated and
defined patches of colour. Cats displaying this coloration are usually female. As we
know, the cat has 38 chromosomes: 36 autosomal and 2 sexual chromosomes. The
male is xy and the female xx. Only chromosome x carries colour; y doesn’t. This
means the male can be xOy red or xoy non-red (i.e. black). The female can be:
–– xOxO red (if both x’s carry orange)
–– xOxo tortoiseshell (if one x carries orange and the other x doesn’t)
–– xoxo black (if both x’s do not carry orange)
The combination xOxo is the only instance (due to the presence of the double x)
in which black and red can appear together.
Tortoiseshell female cats can be black (or another eumelanic colour) and red or
can appear together with white (Fig. 16). When the white is present, the red and
VetBooks.ir
46
M. C. Crosta
Fig. 16 Diluted
tortoiseshell with white
kitten
black are confined into clearly defined and separate patches of colour. The females
that have this particular coat are called tricolour or calico, the North American term.
In presence of the dilution gene, black becomes blue and red becomes cream, giving
rise to the blue-cream coat and, if white is present, to diluted tortoiseshell and white
or diluted calico. In presence of Agouti (A), in the eumelanic areas the tabby pattern
will appear. The term ‘calico’ derives from the Indian city of Calicut, in the Kerala
region, that was a famous port in the sixteenth century thanks to the flourishing
trade between Europe and India. In that city a raw cotton cloth called calico was also
made, that was bleached and then dyed with vibrant colours. This term was later
used in the United States to indicate multi-coloured objects. The distribution and
percentage of colour is defined during the development stage of the embryo. Coats
with grey or white underhair or with tabby stripes on the muzzle or in the red spots
are not allowed. Alongside the classic black-red combination there is also the
chocolate-­cinnamon with red. All of these varieties are admitted with tabby markings (stripes). In the United States, the tortoiseshell or blue and cream tabby is also
called patched tabby or torbie. Tortoiseshell cats are only females. Should this coat
appear in a male specimen, the cat is almost always sterile.
Red
Red is the definition of the coat that has a magnificent golden-red, warm, pure
coloration, evenly distributed from the root to the tip of the hair (Fig. 17). The
standard defines it without any tabby markings (stripes) or lighter spotting. The
Coat Color Genetics
47
VetBooks.ir
Fig. 17 Red and cream
tabby cats
perfect red self (solid colour) is difficult to obtain because the non-agouti gene (a)
that masks the stripes does not have a clean action on pheomelanic colours, as it
does on eumelanic colours. For this reason, sometimes residual marking is visible
on the head and legs, or, in the attempt to eliminate the stripes, coats of a much
too light or washed-out red are obtained. Red self cats without stripes are obtained
only by means of very careful selection. Nose leather and paw pads should be
brick red.
Cream
In presence of dilution genes, red becomes a very soft and delicate pastel cream
(Fig. 17). The colour is evenly distributed from the root to the tip of the hair and
should be as light and homogeneous as possible, without any tabby markings
(stripes), shadowing, light-coloured underhair or darker, pointed areas.
Blue-Cream
In this tortoiseshell cat, the dilution genes soften black into blue and red into cream
(Fig. 16). The colours are perfectly mixed and well distributed, even on the extremities, to create a very light mixture with pastel hues. Like the tortoiseshell, the blue-­
cream cat is only female.
Siamese Pattern
In genetics, when speaking of the Siamese one speaks of coloured points. In fact,
this particular coloration of the extremities is found in many breeds: Siamese, Thai,
Persian (colourpoint) (Fig. 18), Sacred Cat of Birman, Ragdoll, Devon Rex and
Cornish Rex (Si-rex). The gene involved is the one that regulates the intensity of
body colour, i.e. gene C and its mutant alleles called albino alleles.
These alleles are all identified using letter c because they are found on the same
locus (lower case because c is recessive to C, the gene responsible for coat colour
intensity). They all have different suffixes that are the initials of the breeds in which
their action plays a primary role.
48
M. C. Crosta
VetBooks.ir
Fig. 18 Colourpoint cat
•
•
•
•
•
C full colour
cb Burmese
cs Siamese
ca Blue-eyed Albino
c Pink-eyed Albino
All of the alleles of this group are recessive to C but not among themselves; in fact,
between cb (Burmese) and cs (Siamese) there is an incomplete dominance phenomenon. By crossing a Siamese (with light-coloured body and dark tips) with a Burmese
(with shadows of colour, darker on the legs and lighter on the body), one obtains a
Tonkinese (Fig. 19). The Tonkinese shows intermediate features: intensely coloured
tips and body hair which is darker than the Siamese but lighter than the Burmese. Both
(cb and cs), however, are dominant over ca (responsible for the Blue-eyed Albino),
that in turn is dominant over c (Pink-eyed Albino) (C > cs and cb cs > ca > c).
cb Burmese
Albino alleles work by progressively decreasing the pigmentation of the eyes and hair.
In the Burmese, because of the cb gene, black (C) becomes seal (sable or dark sepia)
and the eyes, that are partially depigmented by the gene, tend towards yellow (Fig. 20).
cs Siamese
In the Siamese, the gene cs causes the seal colour to be limited only to the extremities (mask, ears, legs, feet and tail) while the rest of the body is coloured anywhere
from beige to magnolia white. The eyes are deep blue.
Coat Color Genetics
49
VetBooks.ir
Fig. 19 Tonkinese cat
Fig. 20 Burmese cat
c a Blue-Eyed Albino
In the Albino with blue eyes, the lack of pigmentation results in white hair and very
pale blue eyes.
c Pink-Eyed Albino
In the Albino with pink eyes, caused by the presence of the c gene, in addition to the
total lack of pigmentation, the eyes are pink because the iris is transparent and the
retina’s blood vessels become visible.
VetBooks.ir
50
M. C. Crosta
The Siamese gene causes the mutation of the structural gene of the tyrosinase
enzyme, making it especially temperature-sensitive. An increase in temperature,
in fact, deactivates it, and this is why on the body, that is warmer, there is a
decrease in pigmentation (and therefore the coat is paler in colour). At the extremities, that are cooler, there is a larger deposit of pigment that creates the darker
‘point’ effect. Point kittens are born white because in the uterus the temperature
is higher and more constant (38.5 °C), and their colourpoints start showing only a
few days after birth. Climate changes can affect coat coloration too. In fact, cats
living in warm climates are lighter coloured than those that live in cold climates.
The colourpoint in Siamese cats regulates the colour of their body coat: the darker
the points, the darker the rest of the coat (a seal point will have a darker coat compared to a red point). The coat also darkens with age. For this reason, Siamese cats
have a rather short showing career, because the judges prefer more highly contrasting coats.
The Siamese pattern may appear in other cat breeds, such as the Sacred Cat of
Birman, the Devon Rex, the Cornish Rex and others.
White Spotting
Coats with white spots or patches are quite common in nature. The white spots on a
cat’s coat are genetically determined by the S (white spotting) gene and are transmitted as independent entities. This explains why white spotting can be associated
with any basic coat colour. Cats with white spotting are called bicolour and tricolour, and it is common to add ‘and white’ to the cat’s colour name. In this way, a
black cat with white spots becomes ‘black and white’, a red blotched tabby becomes
a ‘red blotched tabby and white’, while the tortoiseshell and white cat is more simply called ‘tricolour’ or ‘calico’. The S gene prevents coat coloration because it does
not allow the melanin granules to settle in the follicles from which the hairs grow.
As the W gene (responsible for the total depigmentation of the hair), S is a dominant
epistatic gene but, unlike W, S does not affect the whole coat but just some patches
of it, and its expression is enhanced by modifier polygenes that can amplify its
action. The white spots show up more intensely if the S is homozygous (SS) compared to its heterozygous state (Ss). For this reason, it is quite common to see cats
with just a few tufts of white hair on the chest and belly, or the extreme opposite
whereby the coats are almost totally white with colour being limited to just a few
areas of the head, back and tail. In fact, due to the variability of these coats, it is
thought that there are various genes (or polygenes) conditioning the expression of
the S gene. The formation of more extensive white areas confines colour into more
distinct and visible spots. In tricolour coats, the spots of red and black are larger
when the proportion of white is higher. Just like the W gene, the S gene seems to be
linked to congenital deafness. It is possible for a white spotted cat to have deafness
in the ear above the blue eye. White spotted coats are classified according to the
percentage of white they contain.
VetBooks.ir
Coat Color Genetics
51
Mitted
The small amount of white (1/4) is limited to the four feet. A white spot is preferred
on the nose and/or between the eyes, while a white line should be present on the
lower part of the body, starting from the throat and ending at the base of the tail.
This coat is typically found in the Ragdoll.
Bicolour
This category includes coats that have a ratio of 2/3 colour to 1/3 white. Colour
should be present on the muzzle (where an upturned ‘V’ is desirable), on the spine,
head, tail and external area of the legs. White is desirable on the chest, belly and the
inside area of the legs (Fig. 21). Also preferable is a white spot on the back, but its
absence is not a penalty. This category allows for coats that show up to 50% white
and 50% colour. Especially appreciated are specimens with a white ‘flame’ on their
muzzle.
Harlequin
This coat has a much higher grade of white spotting compared to colour. Solid
colour, in fact, covers only 1/6 of the coat and is limited to the top of the head, tail
Fig. 21 Bicolour cat
52
M. C. Crosta
VetBooks.ir
Fig. 22 Harlequin cat
and legs. Three or four clearly marked and separate colour spots are desirable on the
back (Fig. 22). Colour spotting is random, but in any case, if located on the back, the
spots cannot be fewer than four. A white flame on the muzzle is highly desirable.
an
V
This category includes coats with colour markings on the head and tail. On the head,
the colour is preferably confined to two large spots separated by a white line between
the ears, while the tail should be evenly coloured down to the base. Not more than
three colour spots on the body are accepted. Coats with more than three colour spots
are considered to be Harlequin. The rest of the body is all-white. This name has
been derived from the Turkish Van, the cat breed that features this coat.
acred Cat of Birman
S
This cat’s coat is long-haired, colourpointed and features white ‘gloves’. The white-­
gloved paws of this cat is the breed’s signature feature, although this peculiar distribution of regular and symmetrical white spots on the four feet has divided genetists
and scholars. Some authors claim that its genotype is similar to that of the colourpoint (cscsll) but with the addition of the S gene (Piebald White Spotting gene).
According to them, the expression of S is conditioned by modifier polygenes that
allow white spots to follow a precise distribution on the four feet. Other authors
VetBooks.ir
Coat Color Genetics
53
instead describe the presence of g (gloves), a recessive autosomic gene that in double dose is capable of confining the white spotting to the extremities. The latter
hypothesis now seems to be the most credited, although it is not precisely understood whether g can be considered a second gene totally different and independent
from S, but capable of changing the expression of the latter (in this case, it would
seem likely that there is a Ssgg genotype in which S codes for spotting and g codes
for the white spotting’s positioning on the feet) or whether it is an allele found on
the same locus as S. Yet others believe that it is a dominant gene with incomplete
penetrance and totally different from S.
Dominant White
White is not a colour but an absence of colour, coded by the dominant, epistatic
gene W. This gene is responsible for the complete depigmentation of the hair
(Fig. 23). It masks the expression of all other colours (epistasis), including white
Fig. 23 White cat
VetBooks.ir
54
M. C. Crosta
spotting and Siamese colourpoint, which means that a white cat can be defined as a
cat of any colour painted white. The progeny of a homozygous white cat will be
entirely white; on the contrary, a heterozygous white cat crossed with a non-white
cat can generate coloured kittens as well. By examining the non-white progeny of
such a cat, one can discover its hidden colour. If one crosses a white male cat with
a red female cat, for example, and this cross produces a tortoiseshell kitten, then the
hidden colour of the male cat will be black.
Sometimes the epistatic effect of this gene is not absolute. Often, a small spot
remains visible on white kittens’ heads, but disappears by adulthood. The W gene is
unfortunately often associated with deafness, because it codes for a degeneration of
the cochlea in the ear and atrophy of the organ of Corti. This genetic defect is congenital and irreversible. The white Oriental, also known as Foreign White, has the
cs gene (Siamese) and the W gene (white), and therefore is a Siamese with the W
gene (cscsW-) and not an albino Siamese from which it differs both by genotype and
by phenotype.
Silver Coats
Cats with silver coats (smoke, shaded, Chinchilla and silver tabby) are perhaps the
most striking and fascinating of them all. In all of these coats, only the tip of the hair
is coloured, while the root section is white. All of these cats share the ‘colour inhibitor’ gene I that prevents the development of pigment in the hair and suppresses its
yellow-greyish banding, which results in a silver coloration effect. Each single hair
of these cats is coloured to varying degrees only at the tip, that can be of any colour:
black, blue, red, tortoiseshell and so on, while the base, closest to the skin, is white.
Even the skin remains normally pigmented. There are many theories regarding the
genesis of silver coats. Until not long ago, the most widely accepted theory was
based on a single gene responsible for this ‘non-pigmentation’, i.e. a mutation of the
colour inhibitor gene I, a dominant autosomic gene that prevents pigment development in the hair (not to be confused with the Albino alleles) probably by limiting the
quantity of pigment destined to the growing hair. The I gene suppresses the yellow-­
greyish bands of the tabby hair and at the same time codes for a pale silver colour at
the base of the hair. In order to differentiate between silver tabby, Chinchilla and
silver shaded, this theory envisaged the intervention of modifier polygenes capable
of regulating the intensity of the I gene and therefore the different proportions
between quantity of coloured hair and silver hair.
Other authors have proposed the presence of another gene, Ch, different and
independent from I. This theory, called the ‘two genes theory’, is based on the
assumption that the I gene erases the yellow-greyish bands and the Ch gene, dominant but independent from I, suppresses ticking and relegates it to the tip of the hair
shaft (tipping).
The most recent theory proposes yet another solution to explain the many questions that arise when analysing the various coats. The presence of the I gene –
colour inhibitor – is confirmed as regards the depigmentation and silver-colouring
VetBooks.ir
Coat Color Genetics
55
of the hair’s base while, to justify the various widths of the underhair, several
polygenes, called Widebanding Genes Wb (undercoat width genes), have been
called into play. The polygenes are capable of regulating band width close to the
base and act in varying degrees (low, medium, high) causing the widening of the
pale band in the Agouti coat. To explain the tipping of the Chinchilla coat, another
recessive gene, the superwide band gene (swb), is assumed to combine with I and
with Wb.
This is the most widely accepted theory at the moment, but due to the complexity
of the matter and to the many issues still to be cleared up, one continues to classify
silver coats based on the varying silver to coloured hair ratio. Accordingly, based on
the width of the coloured part of the hair shaft, called tipping, the following coats
are distinguished.
Smoke
The Smoke is also known as the ‘cat of contrast’ because it has only a very small
white band at the base of the hair in contrast with the very wide band of coloured
tipping. The silver base should be evenly distributed all over the body, head legs and
tail included (Fig. 24). The tipping (from the tip to midway down the hair shaft) is
usually black but can also be blue, red or tortoiseshell. In longhair cats, the contrast
is even more evident. A Smoke Persian, for example, looks entirely black but the
contrast becomes clearly apparent as soon as it moves or when patted.
Shaded
This coat type has tipping (coloured part of the hair shaft) on about 1/3 of the hair
(Fig. 25). The tipping can extend to the muzzle, legs and on the heel and results in a
slightly darker colouring overall compared to the Chinchilla. The coat should not
show tabby markings, dark spots or cream hues. The nose leather is brick red
rimmed with a thin black line. The tips can be of various colours, the most common
being black, although the variations of blue, chocolate, lilac and tortoiseshell are
also admitted.
Fig. 24 Smoke coat
56
M. C. Crosta
VetBooks.ir
Fig. 25 Shaded cat
Fig. 26 Chinchilla cat
Chinchilla
The appearance of the Chinchilla (also called ‘Shell’) is that of a cat with only
light silver tipping and a white underhair. The tipping involves about 1/8 of the
hair (Fig. 26). The chin, chest, belly, inside of the thighs, the underside of the
tail and the hock should be pure white. The head, ears, back, flanks, legs and tail
are slightly shaded due to the presence of the tipping. Silver-shaded and
Chinchilla coats are genetically identical, and can be found together in the same
litter. When the polygenes have an intense action, the result is a Chinchilla, and
when the action is bland the result is a silver shaded. Sometimes it is not easy to
distinguish them. When in doubt, the colour of the heels will give the answer:
silver heels mean silver shaded (Fig. 27), pure white heels mean Chinchilla
(Fig. 28).
VetBooks.ir
Coat Color Genetics
57
Fig. 27 Heels of
Chinchilla cat (lighter
color)
Silver Tabby
This is simply a tabby cat in which the I gene has erased the yellow-greyish bands,
giving place to the white-silver hair to strikingly contrast with the striping above
(Fig. 29).
Cameo
The base of the coat is silver and the tipping is red. Based on the length of the tipping, the cat coats are defined as Smoke Cameo, Shaded Cameo and Shell Cameo.
Golden
This is a special colouring of the coat having a warm apricot coloured underhair and
black tipping. The Golden has the Agouti (A) gene, absence of the I gene (being ii)
and the contemporary presence of wideband polygenes Wb. The genes described in
this chapter are summarized in Table 1.
58
VetBooks.ir
Fig. 28 Heels of silver
shaded cat (darker color)
Fig. 29 Silver tabby cat
M. C. Crosta
Coat Color Genetics
59
VetBooks.ir
The Vibrissae
The Vibrissae, commonly called ‘whiskers’, are very special hairs (Fig. 30). Almost
three times larger and stiffer than normal hairs, they are located three times deeper
into the dermis. They have a sheath of connective tissue rich in elastic fibres and they
are served by a rich array of nerves and blood vessels. In addition to being found on
the cat’s cheeks on the sides of the mouth (12 on each side, in orderly rows), they are
also located above the orbits as well as on the legs at the carpal level. Whiskers move
constantly and stimulate the receptors of the nerve endings. For this reason, they
constitute a powerful information system for monitoring the cat’s immediate surroundings. These highly specialised receptors use afferent neurons to transmit signals to the trigeminal nerve ganglion, and from there to the part of cerebral cortex in
charge of perceiving the somatic-sensorial stimuli, the minimal and almost imperceptible changes in the stimulated vibrissae, as well as all of the highly precise information regarding the extent, direction and duration of this change in status.
Fig. 30 Vibrissae
60
VetBooks.ir
Fig. 31 White cat. The
W gene (dominant
white) masks the
expression of all of the
other genes responsible
for coloration and
colour distribution
Fig. 32 Tabby cat. All
cats are genetically tabby
and possess striping in
their genotype
Fig. 33 Orange cats
(O gene)
M. C. Crosta
VetBooks.ir
Coat Color Genetics
Fig. 34 Tabby kittens.
Agouti is the background
colour against which one
sees the stripes of a tabby
coat
Fig. 35 Siamese cat
(cs gene)
61
VetBooks.ir
62
M. C. Crosta
The position of the vibrissae changes in function of the animal’s activity and
mood: when the cat attacks or is in a position of defence, it will point the vibrissae
towards the rear. The vigil cat, focused on perceiving every single signal, points
them forwards. When instead they are curved forwards and pointing down towards
the ground, they are being used to recognize the ground and are ready to reveal any
pit-holes or other types of unevenness.
The vibrissae pointed forwards to almost embrace the captured victim are being
used to give the exact position of the prey and the direction of its fur or feathers so
that the cat can understand from which end it must swallow it. Vibrissae also play
an important role in defending a cat’s eyes, because they function like eyelashes. It
is enough to touch them lightly and the eyelids will close immediately. This proves
highly useful when hunting, because in its predatory state the cat is concentrated on
the prey, has its pupils totally dilated by adrenalin and therefore finds it difficult to
focus on objects that are very close, such as small branches, bushes, grass or any
other obstacle nearby. Because they just outpast its face, the vibrissae touch these
obstacles first and cause the eyelids to close and defend the eyes.
The tactile functions of the vibrissae have been widely studied and debated. The
vibrissae can act as airflow sensors. They are allegedly capable of detecting – and
therefore of informing the cat about – the smallest vortices of air returned by the
objects it encounters or the weaker air currents created when the air impacts an obstacle. This makes it easy for the cat to move and change position in the dark of night
without bumping into objects. Only such a precise and perfect mechanism can
explain the cat’s incredible skill and precision when hunting at night: with its vibrissae, the cat can acquire an instantaneous and precise perception of its prey and capture it. This happens in blind cats too. A blind or partially sighted cat moves its head
from one side to the other to perceive the ground’s asperities and any obstacles with
its vibrissae. The blind cat lacking vibrissae instead is highly deficient in this respect.
General References
1. Adalsteinnson S. Establishment of equilibrium for the dominant lethal gene for Manx taillessness in cats. Theor Appl Genet. 1980;58:49–53.
2. Affections héréditaires et congénitales des carnivores domestiques, Le point vétérinaire vol 28
N° spécial 1996.
3. Alhaidari Z, Von Tscharner C. Anatomie et physiologie du follicule, pileux chez les carnivores
domestique. Prat Med Chir Anim Comp. 1997;32:181.
4. Alhaidari Z, Olivry T, Ortonne J. Melanocytogenesis and melanogenesis: genetic regulation
and comparative clinical diseases. Vet Dermatol. 1999;7:10.
5. Anderson RE, et al. Plasma lipid abnormalities in the Abyssinian cat with a hereditary rod-­cone
degeneration. Exp Eye Res. 1991;53(3):415–7.
6. Baker HJ, Lindsey JR. Feline GM1 gangliosidosis. Am J Pathol. 1974;74:649–52.
7. Barnett KC, Gurger IH. Autosomal dominant progressive retinal atrophy in Abissinian cats. J
Hered. 1985;76:168–70.
8. Bellhorn RW, Fischer CA. Feline central retinal degeneration. J Am Vet Med Assoc.
1970;157:842–9.
VetBooks.ir
Coat Color Genetics
63
9. Bergsma DR, Brown KS. White fur, blue eyes and deafness in the domestic cat. J Hered.
1971;62:171–85.
10. Biller DS, et al. Polycystyc kidney disease in a family of Persian cats. J Am Vet Med Assoc.
1990;196:1288–90.
11. Bistner ST. Hereditary corneal distrophy in the Manx cat: a preliminary report. Investig
Ophthalmol. 1976;15:15–26.
12. Bland van den Berg P, et al. A suspected lysosomal storage disease in Abyssinian cats. Genetic
and clinical pathological aspects. J S Afr Vet Assoc. 1977;48:195–9.
13. Blaxter A, et al. Periodic muscle weakness in Burmese kittens. Vet Rec. 1986;118(22):619–20.
14. Bosher SK, Hallpike CS. Observations of the histopathological features, development and
pathogenesis of the inner ear degeneration of deaf white cats. Proc R Soc Lond B Biol Sci.
1965;162:147–70.
15. Bosher SK, hallpike CS. Observations of the histogenesis of the inner ear degeneration of the
deaf white cat. J Laryngol Otol. 1966;80:222–35.
16. Bourdeau P, et al. Alopecie hereditaire generalisee feline. Rec Med Vet. 1988;164:17.
17. Boyce JT, et al. Familial renal amyloidosis in Abyssinian cats. Vet Pathol. 1984;21(1):33–8.
18. Boyce JT, et al. Familial renal amyloidosis in Abyssinian cats. Vet Pathol. 1984;21:33–8.
19. Breton RR, Nancy CJ. Feline genetics. Net Pets; 1999.
20. Bridle KH, et al. Tail tip necrosis in two litters of Birman kittens. J Small Anim Pract.
1998;39(2):88–9.
21. Burditt LJ, et al. Biochemical studies on a case of feline mannosidosis. Biochem J.
1980;189:467–73.
22. Carlisle JL. Feline retinal atrophy. Vet Rec. 1981;108:311.
23. Casal M, et al. Congenital hypothricosis with thimic aplasia in nine Birman kittens. ACVIM
abstracts N° 68, Washington, DC; 1993.
24. Centerwall WR, Benirschke K. Male tortoiseshell and calico cats. J Hered. 1973;64:272–8.
25. Chapman VA, Zeiner FN. The anatomy of polydactylism in cats with observations on genetic
control. Anat Rec. 1961;141:205–17.
26. Chew DJ, et al. Renal amyloidosis in related Abyssinian cats. J Am Vet Med. Assoc.
1982;181:140–2.
27. Clark RD. Medical, genetic and behavioral aspects of purebred cats. Fairway: Forum publications Inc; 1992.
28. Collier LL, et al. Ocular manifestations of the Chédiak-Higashi syndrome in four species of
animals. J Am Vet Med Assoc. 1979;175:587–90.
29. Collier LL, et al. A clinical description of dermatosparaxis in a Himalayan cat. Feline Pract.
1980;10(5):25–36.
30. Cooper ML, Pettigrew JD. The retinophthalamic pathways in Siamese cats. J Comp Neurol.
1979;187:313–48.
31. Cooper ML, Blasdel GG. Regional variation in the representation of the visual field in the
visual cortex of the Siamese cat. J Comp Neurol. 1980;193:237–53.
32. Cork LC, et al. The pathology of feline GM2 gangliosidosis. Am J Pathol. 1978;90:723–34.
33. Cork LC, et al. GM2 ganglioside lysosomal storage disease in cats. Science. 1977;196:1014–7.
34. Cotter SM, et al. Hemofilia a in three unrelated cats. J Am Vet Med Assoc. 1978;172:166–8.
35. Counts DF, et al. Dermatosparaxis in a Himalayan cat. Biochemical studies of dermal collagen. J Invest Dermatol. 1980;74:96–9.
36. Creel D, et al. Abnormal retinal projections in cats with Chédiak-Higashi syndrome. Invest
Ophthalmol Vis Sci. 1982;23:798–801.
37. Crowell WA, et al. Polycystic renal disease in related cats. J Am Vet Med Assoc.
1979;175:286–28.
38. Danforth CH. Hereditary of polydactyly in the cat. J Hered. 1947;38:107–12.
39. Davies M, Gill I. Congenital patellar luxation in the cat. Vet Rec. 1987;121:474–5.
40. De Maria R, et al. Beta-galactosidase deficiency in a Korat cat: a new form of feline GM1-­
gangliosidosis. Acta Neuropathol. 1998;96(3):307–14.
VetBooks.ir
64
M. C. Crosta
41. DeForest ME, Basrur PK. Malformations and the Manx syndrome in cats. Can Vet J.
1979;20:304–14.
42. Desnick RJ, et al. In: Desnick RJ, et al., editors. Animal models of inherited metabolic diseases. New York: Liss; 1982. p. 27–65.
43. Di Bartola SP, et al. Pedigree analysis of Abyssinian cats with familial amyloidosis. Am J Vet
Res. 1986;47:2666–8.
44. Donovan A. Postnatal development of the cat retina. Exp Eye Res. 1966;5:249–54.
45. Ehinger B, et al. Photoreceptor degeneration and loss of immunoreactive GABA in the
Abyssinian cat retina. Exp Eye Res. 1991;52(1):17–25.
46. Elverland HH, Mair IWS. Heredity deafness in the cat. An electron microscopic study of the
spiral ganglion. Acta Otolaryngol. 1980;90:360–9.
47. Farrell DF, et al. Feline GM1 gangliosidosis: biochemical and ultrastructural comparisons with
the disease in man. J Neuropathol Exp Neurol. 1973;32:1–18.
48. Flecknell PA, Gruffydd-Jones TJ. Congenital luxation of the patellae in the cat. Feline Pract.
1979;9(3):18–9.
49. Fraser AS. A note on the growth of the rex and angora cats. J Genet. 1953;51:237–42.
50. Freeman LJ. Ehlers-Danlos syndrome in dogs and cats. Semin Vet Med Surg. 1987;2:221.
51. French TW, et al. A bleeding disorder (von Willebrand’s disease) in a Himalayan cat. J Am Vet
Med Assoc. 1987;190:437–9.
52. Gorin MB, et al. Sequence analysis and exclusion of phosducin as the gene for the recessive
retinal degeneration of the Abyssinian cat. Biochim Biophys Acta. 1995;1260(3):323–7.
53. Harpster NK. Cardiovascular diseases of the domestic cat. Adv Vet Sci Comp Med.
1977;21:39–74.
54. Haskins ME, et al. In: Desnick RH, editor. Animal models of inherited metabolic diseases.
New York: Liss; 1982. p. 177–201.
55. Hearing JV. Biochemical control of melanogens and melanosomal organization. J Investig
Dermatol Symp Proc. 1999;4:24–8.
56. Hendy-Ibbs PM. Hairless cats in Great Britain. J Hered. 1984;75:506–7.
57. Hendy-Ibbs PM. Familial feline epibulbar dermoids. Vet Rec. 1985;116:13–4.
58. Hirsch VM, Cunningham JA. Hereditary anomaly of neutrophil granulation in Birman cats.
Am J Vet Res. 1984;45:2170–4.
59. Holbrook KA. Dermatosparaxis in a Himalayan cat. Ultrastructural studies of dermal collagen.
J Invest Dermatol. 1980;74:100–4.
60. Hoskins JD. Congenital defects of the cat. In: Ettinger SJ, Feldman EC, editors. Textbook of
veterinary internal medicine. Philadelphia: Saunders; 1995.
61. Howell JM, Siegel PB. Morphologic effects of the Manx factor incats. J Hered. 1966;57:100–4.
62. Jackson OF. Congenital bone lesions in cats with fold-ears. Bull Feline Advis Bur.
1975;14(4):2–4.
63. Jacobson SG, et al. Rhodopsin levels and rod-mediated function in abysinian cats with hereditary retinal degeneration. Exp Eye Res. 1989;49(5):843–52.
64. James CC, et al. Congenital anomalies of the lower spine and spinal cord in Manx cats. J
Pathol. 1969;97:269–76.
65. Jezyk PF, et al. Alpha-mannosidosis in a persian cat. J Am Vet Med Assoc. 1986;189:1483–5.
66. Jones BR, et al. Preliminary studies on congenital hypothyroidism in a family of Abyssinian
cats. Vet Rec. 1992;131(7):145–8.
67. Johnson CW. The Shaded American Shorthair, 1999 Cat Fanciers’ Association Yearbook, CFA
Inc, New Jersey.
68. Koch H, Walder E. A hereditary junctional mechanobullous disease in the cat. Proc World
Congr Vet Dermatol. 1992;2:111.
69. Kramer JW, et al. The Chédiak-Higashi syndrome of cats. Lab Investig. 1977;36:554–62.
70. “La guide des chats” Selections du Reader’s Digest, 1992.
71. Leipold HW. Congenital defects of the caudal vertebral column and spinal cord in Manx cats.
J Am Vet Med Assoc. 1974;164:520–3.
VetBooks.ir
Coat Color Genetics
65
72. Loxton H. The noble cat, aristocrat of the animal world. New York: Portland House; 1990.
73. Liu S-K. Pathology of feline heart disease. Vet Clin North Am. 1977;7(2):323–39.
74. Livingston ML. A possible hereditary influence in feline urolithiasis. Vet Med Small Anim
Clin. 1965;60:705.
75. Loevy HT. Cytogenic analysis of Siamese cats with cleft palate. J Dent Res. 1974;53:453–6.
76. Loevy HT, Fenyes VL. Spontaneous cleft palate in a family of Siamese cats. Cleft Palate J.
1968;5:57–60.
77. Lomax TD, et al. Tabby pattern alleles of the domestic cat. J Hered. 1988;79(1):21–3.
78. Lorimer. The silver inhibitor gene. Cat Fanciers J.
79. Malik R. Osteochondrodysplasia in Scottish fold cats. Aust Vet J. 1999;77(2):85–92.
80. Martin AH. A congenital defect in the spinal cord of the Manx cat. Vet Pathol. 1971;8:232–9.
81. Mason K. A hereditary disease in the Burmese cats manifested as an episodic weakness with
head nodding and neck ventroflexion. J Am Anim Hosp Assoc. 1988;24:147–51.
82. Muldoon LL, et al. Characterization of the molecular defect in a feline model for type-II
GM2-gangliosidosis (Sandhoff’s disease). Am J Pathol. 1994;144(5):1109–18.
83. Narfstrom K. Hereditary progressive retinal atrophy in the Abyssinian cat. J Hered.
1983;74:273–6.
84. Narfstrom K, et al. Retinal sensitivity in hereditary retinal degeneration in Abyssinian
cats: electrophysiological similarities between man and cat. Br J Ophthalmol. 1989;73(7):
516–21.
85. Neuwelt EA, et al. Characterization of a new model of GM2 gangliosidosis (Sandhoff’s disease) in Korat cats. J Clin Invest. 1985;76(2):482–90.
86. Noden DM, et al. Inherited homeotic midfacial malformations in burmese cats. J Craniofac
Genet Dev Biol Suppl. 1986;2:249–66.
87. Paasch H, Zook BC. The pathogenesis of endocardial fibroelastosis in Burmese cats. Lab
Investig. 1980;42:197–204.
88. Paradis M, Scott DW. Hereditary primary seborrhea oleosa in Persian cats. Feline Pract.
1990;19:17.
89. Patterson DF, Minor RR. Hereditary fragility and hyperextensibility of the skin of cats. Lab
Investig. 1977;37:170–9.
90. Pearson H, et al. Pyloric stenosis and oesophageal dysfunction in the cat. J Small Anim Pract.
1974;15:487–501.
91. Pedersen NC. Feline husbandry. Goleta: American Veterinary Publications Inc; 1991.
92. Prieur DJ, Collier LL. Morphologic basis of inherited coat color dilutions of cats. J Hered.
1981;72:178–82.
93. Prior JE. Luxating patellae in Devon rex cats. Vet Rec. 1985;117(7):154–5.
94. Robinson R. Devon rex: a third rexoid coat mutant in the cat. Genetica. 1969;40:597–9.
95. Robinson R. Expressivity of the Manx gene in cats. J Hered. 1993;84(3):170–2.
96. Robinson R. Genetics for cat breeders. 2nd ed. Oxford: Pergamon Press Ltd; 1987.
97. Robinson R. German rex: a rexoid coat mutant in the cat. Genetica. 1968;39:351–2.
98. Robinson R. The Canadian hairless or Sphinx cat. J Hered. 1973;64:47–8.
99. Robinson R. Oregon rex: a fourth rexoid coat mutant in the cat. Genetica. 1972;43:236–8.
100. Robinson R. The rex mutants of the domestic cat. Genetica. 1971;42:466–8.
101. Rubin LF. Hereditary cataract in Himalayan cats. Feline Pract. 1986;16(4):14–5.
102. Scott DW. Cutaneous asthenia in a cat. Vet Med (SAC). 1974;69:1256.
103. Searle AG, Jude AC. The rex type of coat in the domestic cat. J Genet. 1956;54:506–12.
104. Silson M, Robinson R. Hereditary hydrocephalus in the cat. Vet Rec. 1969;84:477.
105. Simpson J. The white spotting gene: new Zealand Cat Fancy Inc. (NZCF).
106. Sponenberg DP, Graf-Webster E. Hereditary meningoencephalocele in Burmese cats. J
Hered. 1986;77:60.
107. Stebbins KE. Polycystyc disease of the kidney and liver in an adult Persian cat. J Comp
Pathol. 1989;100(3):327–30.
108. Stephen G. Legacy of the cat. San Francisco: Cronicle Books; 1990.
VetBooks.ir
66
M. C. Crosta
109. Turner P, Robinson R. Melaninn inhibitor. A dominant gene in the domestic cat. J Hered.
1980;71:427–8.
110. der Linde V, Sipman JS, et al. Generalized AA-amyloidosis in Siamese and oriental cats. Vet
Immunol Immunopathol. 1997;56(1–2):1–10.
111. Wilkinson GT, Kristensen TS. A hair abnormality in Abyssinian cats. J Small Anim Pract.
1989;30:27.
112. Wright M, Walter S. le livre du chat. Paris: Septimus editios; 1982.
113. Zook BC. The comparative pathology of primary endocardial fibroelastosis in Burmese cats.
Virchow Arch (Pathol Anat). 1981;390:211–27.
114. Zook BC, et al. Encephalocele and other congenital craniofacial anomalies in burmese cats.
Vet Med (SAC). 1983;78:695–701.
VetBooks.ir
Approach to the Feline Patient: General
and Dermatological Examination
Andrew H. Sparkes and Chiara Noli
Abstract
As a naturally solitary species that is both highly territorial and not naturally
social, veterinary visits for the cat and the cat owner can be extremely challenging. The fact that the cat has been removed from its home territory (where it feels
safe) and brought to the clinic (an unfamiliar environment) means that any cat
will naturally experience anxiety, fear, and stress during the visit. For these reasons, it is important that any veterinary visit follows “cat-friendly” principles to
ensure stress is minimized. This will help reduce the severity of stress-induced
changes in laboratory parameters, facilitate an easier clinical examination, and
ensure that owners will be willing to bring their cat back to the clinic when
needed. This chapter will deal with how to perform a general and a dermatological examination, including the description of skin lesions and diagnostic procedures, in the feline patient.
Introduction
As a naturally solitary species that is both highly territorial and not naturally social,
veterinary visits for the cat and the cat owner can be extremely challenging. The
fact that the cat has been removed from its home territory (where it feels safe) and
brought to the clinic (an unfamiliar environment) means that any cat will naturally
experience anxiety, fear, and stress during the visit. For these reasons, it is important
that any veterinary visit follows “cat-friendly” principles to ensure stress is minimized and anxieties are relieved rather than reinforced.
A. H. Sparkes (*)
Simply Feline Veterinary Consultancy, Shaftesbury, UK
C. Noli
Servizi Dermatologici Veterinari, Peveragno, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_3
67
VetBooks.ir
68
A. H. Sparkes and C. Noli
Using cat-friendly principles to minimize stress has numerous benefits. Not only
does it improve the welfare of the feline patient, it also helps to reduce the severity
of stress-induced changes in laboratory parameters, it facilitates an easier clinical
examination, it reduces the risk of human injury from fear-induced feline aggression, and helps to ensure that owners will be willing to bring their cat back to the
clinic when needed.
A more thorough discussion of cat-friendly principles can be found on
International Cat Care’s “Cat Friendly Clinic” web site (see www.catfriendlyclinic.
org), but some of the important issues are covered here.
Before the Cat Arrives
For many owners, the process of taking a cat to the clinic is highly traumatic. They
will have to catch the cat, confine it in a basket, take it away from its natural environment, often transport it in a car, and then bring it into the clinic. Understanding the
implications of veterinary visits for cat owners, and what needs to be done to reduce
the negative impact, will help enormously.
Advising owners on how best to bring the cat to the clinic and helping them
remain calm and relaxed has a very positive effect, both on the owner and the cat.
The cat will be exposed to many stressors such as:
•
•
•
•
•
•
A strange cat basket
An unfamiliar car journey
An unfamiliar environment in the clinic
Strange odors, sights, and noises on the journey and in the clinic
Unfamiliar people and animals, which can be highly threatening
Being handled and examined by unfamiliar people
Suitable cat carriers should be strong, escape proof, and allow easy access for
both the cat, the owner and the clinic staff. Carriers with a large top opening are
usually preferred as they allow easy and gentle lifting of the cat in and out of the
carrier. The carrier should enable the cat to hide if possible, but if it is open on all
sides (e.g., plastic coated wire baskets), then placing a blanket over the carrier to
allow the cat to hide is helpful. Plastic carriers that allow the top half to be removed
completely can be useful as some cats will feel safer remaining in their carrier during a consultation, and most of a clinical examination can be conducted with the cat
in the carrier with the top removed.
Ideally, the carrier should be integrated as “part of the furniture” in the cat’s
home environment. If it is somewhere the cat choses to rest and sleep in on occasions, or if it is somewhere it is fed frequently, the cat will regard it as part of its
territory rather than seeing it as a clue that a stressful journey is ahead, if it only
comes out for veterinary visits. Ensuring that some of the cat’s usual bedding is used
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
69
in the carrier during the visit will also be reassuring to the cat as this will contain
the odors the cat associates with its home territory. In addition, the use of synthetic
feline facial pheromone spray or wipes in the carrier and/or on the bedding may be
helpful. Asking the owner to bring extra bedding is also a good idea in case the cat
soils the basket with feces or urine.
During a car journey, ensuring the carrier is restrained securely (eg, in a foot
well) and will not move during the journey is important. Driving calmly will be
helpful and if necessary cover the basket with a blanket to ensure the cat is able
to hide.
With cats that are known to repeatedly become highly anxious and aroused during a veterinary visit and during the journey to the clinic, consideration can be given
to the use of anxiolytic drugs such as gabapentin [1, 2]. While not recommended
for routine use, there are undoubtedly some cats that will benefit from such an
approach.
The Waiting Room
A well-designed waiting room with cat-friendly staff is important. The aim should
be to create a calm and non-threatening environment for the cat to wait in so that
anxiety is reduced rather than heightened. An atmosphere that reassures owners that
the clinic is staffed by people who care about both them and their cats will also help
to create a positive impression.
The waiting room should be designed and used in a way that minimizes the
threats cats may feel (visual, aural, olfactory, etc.). Ideally, a clinic would have a
separate waiting room for cats, but if this is not possible, consider physically separating the waiting room into two different areas for dogs and cats. Appropriate walls
or barriers should be used to ensure visual contact is avoided between dogs and cats
(Fig. 1), and measures should be taken to avoid having barking or noisy dogs in the
waiting room (e.g., getting noisy dogs to wait outside).
Fig. 1 Having a separate
waiting area for cats and
cat owners that is quiet,
and where cats cannot see
dogs helps to reduce stress
during veterinary visits
VetBooks.ir
70
A. H. Sparkes and C. Noli
The location and size of the cat waiting area should be appropriate for the
clinic, and thought should be given to the route that cats take into and out of it.
The cats should encounter minimal human and animal traffic while in the waiting
area. The value of a feline-only area is greatly compromised if cats have to pass
through a noisy area or pass by dogs to get to the consulting room. Having the cat
waiting area adjacent to a cat consulting room can help overcome some of these
problems.
Other important considerations for the feline waiting area include:
• Having a low reception desk, or a wide shelf in front of the reception desk where
owners are encouraged to place cat baskets (above the head height of most dogs).
This helps to reduce anxiety as cats feel more threatened when at floor level;
• Prevent or reduce any noises from the consultation rooms reaching the waiting
area;
• Ensure dogs are kept away from cat carriers, and reinforce this by asking dog
clients to be considerate of cats in the waiting area;
• Try to ensure cats are not left to wait for excessive periods of time in the waiting
room, but are able to move to the consulting room as quickly as possible;
• Direct visual contact with other cats can also be very threatening. This can be
overcome in many ways such as erecting small partitions between seats to separate cats in the waiting area, or providing clean blankets or towels to cover the
cat’s carrier;
• Cats feel insecure if placed at floor level – having shelves, tables or chairs to
place cat carriers on so they are raised up is very useful (Fig. 2). These should
ideally be about 1.20 m from the ground and have partitions (or use covers) so
that cats are not confronted with each other;
• Using a plug-in synthetic feline facial pheromone diffuser (Feliway, Ceva Animal
Health) may also be of benefit in the environment.
Fig. 2 Having raised
tables or shelves above
floor level for owners to
place the cat basket on
while in the waiting room
is another good way to
provide reassurance to cats
and help reduce anxiety
Approach to the Feline Patient: General and Dermatological Examination
71
VetBooks.ir
The Consultation Room
Where possible, a clinic should use a dedicated feline consulting room, free from
the odor of dogs and other animals. For dermatologic examinations the room should
be well lit, but with the ability to darken the room if needed (for example for evaluation with a Wood’s Lamp). There should also be access to an illuminated medical
magnifier.
Consideration should be given to allowing the cat to wander freely in the consulting room if it chooses, and so it is important there are no cupboards or furniture in
the room that the cat could hide under, or small gaps that would make it difficult to
retrieve the cat. The consulting room table should also have a clean non-slip surface
so that the cat is able to grip well – this can be achieved with a rubber mat or perhaps
a clean thick towel or blanket.
The use of synthetic feline facial pheromone sprays and diffusers in the consulting room may help to encourage a more relaxed atmosphere, but this is not a substitute for good empathetic handling techniques.
The Consultation Process
The aim of the consultation process should be to obtain a full history, undertake a
full physical examination, and consider what further actions or investigations may be
required in conjunction with the owner, while ensuring the cat remains as stress-­free as
possible. Irrespective of the suspected cause of the skin disease, a full history and full
clinical examination should never be overlooked as there may be concurrent disease
present and/or a systemic cause of the skin condition. A properly conducted dermatologic consultation, including ancillary tests, usually requires 45–60 minutes. About
20 minutes are dedicated to collecting and recording the signalment and history, examination of the patient requires about 10 minutes and the ancillary tests and discussions
with the owner each require about 15 minutes. The times are indicative only and vary
depending on the nature of the problem and the communicative ability of the owner.
The principles of “cat-friendly” handling should be adhered to at all times – see
the AAFP/ISFM Feline-Friendly Handling Guidelines [3] – and the cat should be
given time to acclimatize to this unfamiliar environment.
History Taking
Collecting and reviewing information on the medical and surgical history of the cat
is a part of the routine healthcare examination. The history should be collected, as
far as possible, in a systematic way – using a clinical history form is a valuable way
of obtaining standardized data for all patients (Figs. 3, 4 and 5).
A. H. Sparkes and C. Noli
VetBooks.ir
72
Fig. 3 Example of a feline clinical history form. Copies of this form are freely available from
www.catcare4life.org
73
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
Fig. 4 Example of a feline nutritional history form. Copies of this form are freely available from
www.catcare4life.org
A. H. Sparkes and C. Noli
VetBooks.ir
74
Fig. 5 Example of a feline physical examination form. Copies of this form are freely available
from www.catcare4life.org
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
75
Clinical history and/or health questionnaires (e.g., including behavior, mobility,
routine prophylactic therapy, and general health) can be given to owners to fill out,
as far as possible, before bringing their cat to the clinic or while in the waiting room
before the consultation. The assistance of a nurse or member of support staff may
be valuable, but collecting such information before the consultation itself helps to
streamline the process and gather all relevant information.
In particular for dermatological consultations it is highly desirable to record
all the data collected on a special dermatological clinical record form. This form
should be divided into sections for the signalment, history, clinical examination, list
of differential diagnoses, ancillary tests, definitive diagnosis, therapy and follow-up
(Fig. 6).
Many of the issues may be obvious, and may be a part of the existing medical
record if the cat is a long-standing patient at the clinic. However, it is important to
remember that some owners take their cat to more than one veterinary clinic, so
other relevant problems the cat may have suffered should not be overlooked. Even
when an accurate history is known, it is still important to consider:
• Any current medications (prescribed by the clinic or obtained elsewhere)
• Any non-prescription medications the owner may be using (e.g., nutritional supplements, parasiticides, alternative medications, etc.)
• Lifestyle (indoors, outdoors, other animals in the house, etc.)
In particular, questions regarding the skin disease for which the cat is presented
should include:
•
•
•
•
age of onset/duration of the problem;
seasonality;
initial site and lesion type and its modification during the course of the disease;
severity and localization of pruritus, if present (Tables 1 and 2).
Reviewing the history during the clinical examination is an ideal opportunity to
open the cat carrier and allow the cat time to come out voluntarily and explore the
room. This helps acclimatize the cat to the environment and helps reduce stress during the subsequent examination.
History-taking should always include the use of open-ended questions such as:
• “How has Fluffy been doing since the last visit?”
• “Have you noticed any change in his appetite recently?”
• “Has there been any change in Fluffy’s stool consistency?”
These are always better than leading questions such as:
• “Have you seen any diarrhea?”
• “Has he been eating more recently?”
VetBooks.ir
76
Fig. 6 Example of a feline dermatological examination history form
A. H. Sparkes and C. Noli
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
Fig.6 (continued)
77
VetBooks.ir
78
Table 1 Feline skin diseases
and potential severity of
pruritus
Table 2 Feline skin diseases
and most frequent
localization of pruritus
A. H. Sparkes and C. Noli
Potential severity of pruritus
Absent
Non-inflammatory alopecia
Demodicosis (D. cati, uncomplicated)
Dermatophytosis (uncomplicated)
Moderate
Feline atopic syndrome (uncomplicated)
Adverse reaction to food (moderate severity)
Bacterial infection
Malassezia infection
Demodicosis by D. gatoi or by D. cati
Cheyletiellosis
Severe
Severe food allergy
Severe Malassezia infection
Notoedric mange
Most frequent localization of pruritus
Dorsum
Flea bite allergy
Cheyletiellosis
Psychogenic (licking)
Other allergies
Head
Otodectic mange
Adverse reaction to food
Notoedric mange
Neck
Flea bite allergy
Adverse reaction to food
Idiopathic neck lesion (consider welfare issues)
Abdomen (self-induced alopecia in cats from licking)
Feline atopic syndrome
Flea bite allergy
Adverse reaction to food
Flea infestation
Cheyletiellosis
Psychogenic
Importantly, a full history should also include a good nutritional assessment
(Fig. 4), evaluating the cat’s diet, lifestyle, feeding habits, etc. It is important that
this is as comprehensive as possible and covers everything the cat has access to.
The cat’s behavior and environment should not be overlooked. This will include
whether the cat has free access outdoors, what other animals it regularly has contact
with and whether the cat is known to hunt. It is also important to consider the potential interplay between many medical and behavioral issues, including dermatoses
(e.g., psychogenic dermatoses).
Approach to the Feline Patient: General and Dermatological Examination
79
VetBooks.ir
Physical Examination
Patience, gentleness, and empathy are vital characteristics with cats in the consultation room. Even with the best environment and approach, some cats will remain
very anxious and a full physical examination may not always be possible at the first
attempt. Be prepared to take additional time if needed, and in some cases consider
scheduling another appointment, or hospitalizing the cat if necessary.
As with the history taking, using a standardized form for physical examination
and additional forms for special investigations such as dermatological examinations
will be highly valuable (Figs. 5 and 6). Using a standardized form will ensure that
the physical examination is performed systematically and that nothing is missed.
This can be particularly important in cats as the order of events during the examination may have to be flexible and adapted to the needs of the individual cat (see
below).
Important considerations during the physical examination include:
• Don’t ever rush when examining a cat – taking a little extra time to do things
slowly and at the cat’s pace will be much more rewarding and less stressful.
• Always try to let the cat come out of its carrier by itself.
• Be flexible and let the cat choose – allowing the cat some control through exercising choice is a key method to reduce anxiety. The key is to find out and understand what makes the cat more relaxed and adapt the physical examination to suit
the individual cat. Some cats may be happier on their owner’s lap, others on the
floor. Some may enjoy looking out of a window, while others prefer to stay sitting in their carriers or even hiding under a blanket. Try to be as adaptable as
possible, be gentle, and take your time.
• Give the cat plenty of fuss and attention if that is what it likes, talk gently and aim
to complete the majority of the physical examination without the cat realizing
you are doing anything more than just stroking it.
• Providing some treats, if the cat will eat them, may also help to distract the cat.
• Sitting with the cat on the floor often helps and can make handling much
easier.
• Some cats prefer to lie down, while others prefer to stand – try to do as much as
possible with the cat in its preferred position.
• Always use the minimal amount of restraint necessary – any form of overt or
heavy restraint will signal danger to the cat and escalate anxiety.
• Where needed, split the examination into short sections, and in between allow
the cat to rest, change position, or wander round the room – give the cat a short
break as soon as it starts to get restless.
• As sustained eye contact with a cat can be perceived as threatening by the cat,
avoid direct eye contact where possible, and perform as much of the examination
as possible with the cat facing away from you (Fig. 7).
• Be aware that older cats often suffer from osteoarthritis, which may make handling uncomfortable or painful.
• Perform more invasive examinations (such as taking the cat’s temperature, where
necessary) to last.
VetBooks.ir
80
A. H. Sparkes and C. Noli
Fig. 7 The physical
examination should be
conducted gently and
empathetically. Conducting
much of the examination
from behind the cat avoids
direct eye contact which
cats often perceive as
threatening
In particular, for a dermatological examination, the following should be evaluated before any manipulation:
•
•
•
•
•
Nutritional status
Coat luster
Thickness of the coat
Any odors
Localization of any obvious lesions
When possible the coat and skin should be inspected systematically. The authors
normally follow a precise sequence so as not to forget any part of the body.
1. Rear of the animal
–– The area at the base of the tail is examined, extending forward along the dorsum to the neck.
–– The areas under the tail, anus, and perianal and perivulvar (females) skin are
inspected.
–– The hind legs are examined, checking for any linear granulomas.
–– The hind feet are checked, examining all the interdigital spaces from underneath and on top. The nail beds are examined and all the nails are exposed and
checked.
2. In recumbency
–– The medial aspect of the hind limbs and the inguinal and abdominal areas are
examined. The external genitalia are inspected, including exteriorizing the
penis and opening the vulva.
–– The sternal area, axillae, and medial aspect of the front limbs are examined.
3. Side of the animal
–– The lateral thorax and neck are examined and then the front leg and foot.
–– Examination of the appearance and odor of the external ear.
–– Repeat the examination from the other side.
4. Finally, the patient is examined from the front
–– The head is inspected, including opening the mouth and examination of the
conjunctivae.
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
81
The whole process should be performed gently and quickly, in order not to stress
the patient.
In exceptional cases, some cats are so fearful that a full examination is not
achievable even with the most patient of handling. This is rare, but heavy restraint
(scruffing the cat and pinning it to the table) will only make the experience much
worse for the cat. In such cases, consider using chemical restraint to facilitate the
examination.
Cats should be weighed at every clinic visit, and at least once or twice yearly.
The percentage weight change should be calculated at each visit and trends noted.
Human pediatric or feline-specific accurate electronic scales should be used if possible for optimum accuracy.
Skin lesions and their localization and distribution should be recorded for future
reference. These include:
Macule A non-raised area of a color different to that of the surrounding skin.
Hyperpigmented macules on the skin and mucosae of orange cats represent lentigo
simplex (Fig. 8). Erythematous macules may be derived from peripheral vasodilation (as it occurs in many inflammatory skin diseases) or from hemorrhage (petecchiae). An extensive area of erythema is called erythroderma. Depigmented macules
are typical of vitiligo in Siamese cats.
Papule A small, raised, erythematous lesion, it represents accumulation of inflammatory cells within the skin. Papules are typical, e.g., of the initial phases of eosinophilic granulomas. Papules are also a feature of parasitic skin diseases (mosquito-bite
hypersensitivity, Fig. 9) and xanthomas.
Pustule An accumulation of inflammatory cells (pus) within or just under the epidermis. In cats, pustules are very rare and most frequently seen with pemphigus
foliaceus (Fig. 10).
Fig. 8 Brown maculae
(lentigo simplex) on the
oral mucosae of a red cat
VetBooks.ir
82
A. H. Sparkes and C. Noli
Fig. 9 Small papules and
erosions on the pinna of a
cat with mosquito bite
hypersensitivity
Fig. 10 A pustule on the
footpad of a cat with
pemphigus foliaceus
Vesicle An accumulation of clear or hemorrhagic fluid within or just under the
epidermis, a rare lesion often caused by autoimmune skin diseases.
Cysts Non-neoplastic well-circumscribed accumulations of liquid or keratin.
Multiple apocrine cysts, containing clear fluid, are observed in Persian cats (Fig. 11).
Approach to the Feline Patient: General and Dermatological Examination
83
VetBooks.ir
Fig. 11 Apocrine cysts on
the muzzle of a Persian cat
Fig. 12 Numerous
nodules on the head of a
cat with feline progressive
histiocytosis
Nodule A raised protuberance caused by the infiltration or proliferation of cells and/
or excessive connective stroma. Nodules are seen in bacterial disease (e.g., abscess),
fungal infection (e.g., deep mycosis or dermatophyte mycetoma), sterile reactions
(injection site granulomas, feline progressive histiocytosis, Fig. 12), or neoplasia.
Plaque A firm, raised area with a flattened surface, e.g., eosinophilic plaque
(Fig. 13).
Wheal Raised well-circumscribed lesion consisting of edema of the superficial
dermis. Wheals have an acute onset (a few hours) and tend to resolve quickly (over
VetBooks.ir
84
A. H. Sparkes and C. Noli
Fig. 13 Typical aspect of
a eosinophylic plaque in an
allergic cat
Fig. 14 Angioedema of
the head of a cat
a few hours or 1 day). Wheals are a manifestation of a type 1 hypersensitivity reaction (immediate or anaphylactic) and are seen in reaction to intradermal allergen
tests. Angioedema is edema extending to the deeper tissue and involving a larger
area of the body (esp. the head, Fig. 14).
Comedones Commonly referred to as “blackheads”, they represent an accumulation of keratin in the infundibulum of the hair follicle. Comedones in cats are seen
on the chin in feline acne (Fig. 15).
Crust The accumulation of dried exudate (Fig. 16) or blood. The color depends on the
material from which they were formed (blood = brown, pus = yellow). The eschar
(Fig. 17) is a particular type of crust that contains dermal collagen fibers and is thus
strongly anchored to the body (i.e., it cannot be easily pulled). Eschars are typically seen
in cats in case of idiopathic ulcerative neck lesion and feline perforating dermatitis.
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
Fig. 15 Comedones and
furuncolosis on the chin of
a cat affected with acne
Fig. 16 Several yellow
crusts (dry pus) on the
pinna of a cat affected with
pemphigus foliaceus
Fig. 17 An eschar on the
neck of a cat affected with
feline idiopathic ulcerative
dermatitis
85
VetBooks.ir
86
A. H. Sparkes and C. Noli
Fig. 18 Dry large flaques
of exfoliation in a cat
affected with
paraneoplastic thymoma-­
associated exfoliative
dermatitis
Fig. 19 Self-inflicted
excoriations and
ulcerations in a cat with
adverse food reactions
Scale Dry accumulations of layers of the stratum corneum commonly called dandruff
(Fig. 18). The presence of scales in cats is usually associated with dermatophytosis,
sebaceous adenitis, or paraneoplastic diseases (feline exfoliative dermatitis due to
thymoma).
Excoriation A self-induced lesion including ulceration and crusts, resulting from
scratching and/or biting (Fig. 19).
Erosion A loss of epidermis down to the level of the basement membrane but leaving the dermis intact. Erosions are seen in some autoimmune diseases (e.g., pemphigus complex and diseases inducing dermo-epidermal separation) and in early cases
of eosinophilic plaque (because of the abrasive action of the feline tongue).
Ulcer Tissue loss involving the epidermis and underlying tissues (dermis, less frequently subcutis). Examples of ulcers in cats are deep bacterial (e.g., atypical myco-
Approach to the Feline Patient: General and Dermatological Examination
87
VetBooks.ir
Fig. 20 Lesion of lip
(indolent) ulcer
Fig. 21 Self-inflicted
alopecia on the abdomen
due to licking
bacteria) or fungal (e.g., sporotrichosis) infections, for squamous cell carcinoma,
idiopathic neck ulcer and lip (indolent) ulcer (Fig. 20).
Draining tract An opening in the tissue releasing exudate produced by a deeper
inflammatory process (dermis or subcutis). Fistulization of abscesses or other
inflammatory foci (sterile panniculitis, foreign body granuloma, etc.) permits the
drainage of pus and the eventual expulsion of etiological agents, foreign bodies, or
necrotic material.
Alopecia Can be used to describe both the complete loss of hair over one or more
areas of the body and hypotrichosis, meaning thinning of the hair coat. It is important to differentiate alopecia caused by the loss of the hair together with the root
from the loss of part of the hair shaft only. In cases of the loss of the hair root, for
example, in endocrine or paraneoplastic alopecia, the hair at the periphery of the
lesion can be easily epilated with traction. In the case of broken hair (e.g., self-­
induced alopecia, Fig. 21), the small remaining ends of the hair can be felt or seen
VetBooks.ir
88
A. H. Sparkes and C. Noli
with a magnifying lens as they leave the follicular ostia. The hair surrounding the
areas of alopecia resists epilation.
Diagnostic Investigations
There are a number of diagnostic tests that, although mostly very straightforward,
are extremely valuable in the diagnosis of various dermatoses. Again, it is important
to consider appropriate chemical (sedation) for the cat in order to facilitate these
tests wherever necessary. It is far less stressful to use appropriate chemical restraint
than to struggle with heavy physical restraint of an anxious or fearful cat.
Trichogram
A trichogram involves microscopic examination of hairs (tip, shaft, and root) – ideally
around 20–30 hairs are plucked and then examined. The preferred instrument to remove
the hair is a pair of mosquito hemostats (preferably Klemmer) covered by small rubber
or plastic tubing (to obtain even pressure and avoid causing artifacts by damaging the
hair). In this way, samples from all stages in the growth cycle will be obtained rather
than only resting-phase hairs. The hairs should be plucked in the direction of the hair
growth to avoid fracturing them at the base. The hairs can be secured on a microscope
slide either by placing them in mineral oil with a coverslip placed on top or using adhesive acetate tape. Examination is performed under 40× and 100× magnification.
The hair tips can be examined to determine whether there is pruritus (traumatic
epilation) or spontaneous epilation. With traumatic hair loss the tips of the hairs will
be broken and the usual slender tapering tips will be lost.
The hair roots can be examined to determine if the hairs are in anagen or telogen
and see if there is normal hair cycling. Most should be in telogen (rough, spear-­shaped
bulb), with fewer in anagen (expanded bulb, may appear fringed, often pigmented,
may appear club-shaped). In shorthair cats around 90% of hairs will be in telogen.
Hair shafts should also be examined for abnormalities including the presence of
ectoparasites (Demodex cati), ectoparasite eggs (Felicola subrostratus, Chyeletiella
spp.), and/or dermatophytes. Large adhesions of keratin on the hair shafts, called
follicular casts, are seen in sebaceous adenitis.
Skin Scraping
A small window of skin is clipped if necessary. A small amount of mineral oil can
be put on the skin surface to facilitate the skin scraping and a blunted scalpel blade
or a Volkmann spoon (diameter 5–6 mm) is held perpendicular to the skin surface,
which is scraped in the direction of the hair growth using moderate pressure.
For superficial parasites such as Notoedres cati and Demodex gatoi, scraping
should not be so deep to cause capillary oozing of blood. The collected material can
be smeared on a slide for examination.
Deep skin scraping involves repeated scraping at the same site until there is capillary oozing of blood. Pinching the skin prior to scraping to “squeeze” contents out
from the follicles may also facilitate collection of follicular material and follicular
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
89
Demodex mites. In untreated patients with demodicosis, the number of mites detected
is usually very high and hence it is rarely necessary to collect more than 2 or 3 samples
and trichoscopy may be preferable over skin scrapings. When monitoring therapeutic
success the mite numbers are low and numerous deep skin scrapings are necessary.
Examination of skin scrapes is performed under 40× to 400×, but initial examination should always be done at 40×. If there is heavy keratinous debris, taking “dry”
skin scrapes and suspending the collected material in 10–20% potassium hydroxide
which is then left for 20–30 minutes before examination under a coverslip may
enhance visualization through “clearing” of the keratinous debris.
ape Strip Test (“Scotch Tape” or “Acetate Tape” Test)
T
This test allows collection of superficial skin parasites, hairs, and yeasts. A 5–8 cm
strip of clear sticky tape is repeatedly applied to the lesion or area of skin of interest.
The skin can be clipped prior to performing the test if necessary. The tape is then
applied (stuck) to a microscope slide. The free ends can be wrapped around the slide
to help anchor it.
The preparation can be stained if necessary (e.g., looking for Malassezia) by
applying a drop of suitable stain (e.g., the “blue” Diff-Quik stain) to the slide before
applying the sticky tape. The slide is examined under 40× to 400× magnification.
Coat Brushings
Coat brushings are particularly helpful to look for fleas, but may also provide evidence of other superficial parasites. The cat is placed on a large sheet of white paper
and is brushed vigorously both with and against the growth of the hair. Scale and
debris are collected, are examined macroscopically, and can be placed on a slide and
examined in mineral oil or a stain such as lactophenol cotton blue. Combing with a
flea comb may also be a useful part of the procedure.
ood’s Lamp Illumination
W
Wood’s lamp illumination is used to examine the hair coat (or collected hairs) under
ultraviolet light to look for spontaneous fluorescence that is often associated with
Microsporum canis infection. For optimal results, it is important to use the Wood’s
lamp in a dark room, and to allow 30–60 minutes for the eyes to adapt to the low
light conditions. More information regarding this technique is presented in the
chapter dealing with dermatophytosis (Chapter, Dermatophytosis).
Cytology
Several techniques have been developed to obtain material for a cytological
examination.
Fine Needle Aspiration This technique is used for raised lesions, nodules, or
accessible lymph nodes. A 21G (gray) needle is inserted into the center of the nodule and connected to a 5 or 10 ml syringe. While the needle is inside the mass several 1–2 ml aspirates are made, changing the needle position (angulation) without
withdrawing the needle from the lesion. The suction is completely released before
VetBooks.ir
90
A. H. Sparkes and C. Noli
withdrawing the needle. If this has been done correctly, the plunger should return to
0 ml and the cells will be located in the needle lumen. If the plunger fails to return
to zero, air has entered the syringe and the procedure should be repeated because the
cells are in the cone of the syringe and difficult to remove. The needle is then
removed from the cone and the syringe filled with air, reconnected to the needle and
the cells are “sprayed” onto a microscope slide. If the sample is liquid, a smear is
made, similar to that used for blood. If the material is solid, the material is spread
by placing a second slight over the sample, applying little or no pressure.
Fine Needle Insertion A 24G needle is inserted into the mass and its angle
changed, without connection to a syringe. The needle is then removed from the
lesion, connected to a syringe filled with air and the samples are sprayed onto a
slide and spread as described above. This technique is particularly indicated for
lymph nodes, for very small lesions or when excessive blood is obtained with
aspiration.
Impression Smear Impression smears are used for exudative lesions, superficial
oily accumulations, pustules, crusts, or biopsy specimens cut in half. The slide is
placed several times lightly onto the lesion or oily area. To sample a pustule or a
crust, the lesion is opened with a 24G needle and a slide is applied to the drop of pus
that comes out. Impression smears have the advantage of not deforming the cells but
often result in a sample that is too thick. In such cases, search around the margins of
the slide for a monolayer of cells.
Superficial Skin Scrapings As previously described, Malassezia can be demonstrated with a very superficial skin scraping of seborrheic skin by using a number 10
or 20 scalpel blade. The material is spread onto a microscope slide using the blade,
fixed using a flame and stained with a standard stain.
Sampling with a Cotton Bud This technique is useful for collecting samples from
draining tracts, interdigital spaces, claw folds, and external ear canals. The sample
is applied to a slide by gently rolling the cotton bud.
Skin Biopsy
Skin biopsies are sometimes needed for investigation and diagnosis of dermatoses.
If the condition of the patient permits, it is better to perform the biopsy after
1–2 weeks of antibiotic therapy. This removes any secondary infection that can
complicate the interpretation of the biopsy. Preferred antibiotics include cephalexin (20–30 mg/kg orally twice daily), cephadroxil (20–30 mg orally twice daily),
or amoxicillin-clavulanate (20–25 mg orally twice daily). To avoid secondary
infection and scarring, the antibiotic can be continued for 1 week after the biopsy.
If the patient has been receiving glucocorticoid therapy and the condition of the
patient permits, then the biopsy should be delayed until 15–20 days after treatment
discontinuation or longer if long-acting depo-injections have been used.
VetBooks.ir
Approach to the Feline Patient: General and Dermatological Examination
91
Local anesthesia would be preferable, given that the procedure is minor and
rapid and requires only one or two sutures, however in cats this is possible only if
they are extremely quiet and when biopsies are taken from the trunk. If using local
anesthesia, one should remember that no more than 1 ml of 2% lidocaine should be
injected in cats, due to risk of cardiac toxicity. If multiple biopsies are necessary,
lidocaine can be diluted 1:1 with saline, so that 2 ml of 1% lidocaine are obtained
and can be used for up to 4 biopsies. In the majority of cases, however, general
anesthesia is used.
In general, collecting several biopsies from representative lesions will facilitate the diagnosis. Wherever possible, early lesions, such as papules and pustules
should be biopsied, and later evolution of these, such as ulcers and crusts should be
avoided, however, if there is a range of lesions, biopysing all is prudent.
Prior to biopsy, lesions may be gently clipped, but it is preferable not to clean the
skin as this may remove valuable diagnostic material. Disposable punch biopsies
are usually the preferred method of biopsy collection. Biopsies from the edge of
lesions, including adjacent apparently normal-looking skin, should be obtained with
the elliptical excision biopsy technique.
The sample should be put in 10% fresh formalin and accompanied by a full clinical history. The pathologist should be informed of the signalment (age and breed),
clinical signs, description and site of the lesions, and duration and evolution of the
disease. Any treatment/medication, its duration and period of suspension should be
included. Biopsies from different sites should be submitted in separate, numbered
containers with a description in the history indicating the site and type of lesion for
each biopsy.
References
1. Pankratz KE, Ferris KK, Griffith EH, et al. Use of single-dose oral gabapentin to attenuate fear
responses in cage-trap confined community cats: a double- blind, placebo-controlled field trial.
J Feline Med Surg. 2018;20:535–43.
2. Van Haaften KA, Eichstadt Forsythe LR, Stelow EA, Bain MJ. Effects of a single pre-­
appointment dose of gabapentin on signs of stress in cats during transportation and veterinary
examination. J Am Vet Med Assoc. 2017;251:1175–81.
3. Rodin I, Sundhal E, Carney H, et al. AAFP and ISFM Feline-Friendly Handling Guidelines. J
Feline Med Surg. 2011;13:364–75.
VetBooks.ir
Part II
Problem Oriented Approach to…:
VetBooks.ir
Alopecia
Silvia Colombo
Abstract
Alopecia, either spontaneous or self-induced, is a common presenting sign in
cats. Definitions of alopecia and hypotrichosis and the clinical features of alopecia are given at the beginning of this chapter, followed by pathogenesis of the
different types of alopecia. Clinical presentations of alopecia and its preferential localization in selected feline diseases are described, together with useful
diagnostic hints coming from signalment and history. The diagnostic approach
to alopecia implies the correct differentiation of the pathogenetic mechanisms
underlying the clinical signs, which can be obtained by collecting history, examining the cat, and performing a microscopic examination of the hair. This is, in
cats, a very important diagnostic test, which should always be performed at the
beginning of the consultation, in order to differentiate spontaneous from self-­
induced alopecia. Dermatophytosis is very common in cats, and diagnostic tests
to diagnose or rule out this disease should be carried out in all cases presenting
with alopecia.
Definitions
Alopecia simply means hair loss. The word alopecia is derived from the ancient
Greek word ἀλώπηξ (alṓpēx), which means fox. The term “alopecia” was used at
that time to describe fox mange.
Hypotrichosis means that there is less than normal amount of hair (from the
ancient Greek words υπο, below, and θριξ, hair), and this term is sometimes used
as a synonym of partial alopecia. Although the exact meaning of these two terms
is very similar, if not identical, the term hypotrichosis is preferred when there is a
S. Colombo (*)
Servizi Dermatologici Veterinari, Legnano, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_4
95
VetBooks.ir
96
S. Colombo
Fig. 1 Congenital
hypotrichosis in a domestic
short-haired kitten
congenital deficiency of hair in both human and veterinary dermatology publications (Fig. 1) [1]. Strictly speaking, hypotrichosis should be used as a synonym of
congenital alopecia.
Alopecia can be classified depending on severity (partial or complete), distribution
(focal, multifocal, generalized, symmetrical), localization, and pathogenesis. Partial
alopecia means that there is less than normal amount of hair, while complete alopecia
describes absence of hair. Focal alopecia, occasionally also called localized alopecia,
refers to a single patch of alopecia anywhere on the body (Fig. 2). If more patches
are present, alopecia is defined as multifocal. Focal or multifocal alopecia in cats is a
clinical presentation commonly observed in cases of dermatophytosis (Fig. 3). When
a whole region of the body is involved, alopecia is described as diffuse or generalized.
Diffuse alopecia may be symmetrical, when both sides of the body are equally affected.
Generalized alopecia is normal in hypotrichotic breeds, such as the Sphynx cat [2].
Pathogenesis
In cats, the most useful classification of alopecia from a diagnostic point of view
is the one based on pathogenesis. Alopecia can be spontaneous, when the hair falls
off, or self-induced, when the cat actively removes the hair by continuous licking.
Spontaneous alopecia occurs as a consequence of two main pathogenetic mechanisms. When inflammation or infection targets the hair follicle and/or the hair shaft, the
latter undergoes damage and falls off (Fig. 4). Hair may also be absent because the hair
follicle is dysplastic or atrophic, thus not able to produce a normal hair shaft (Table 1).
VetBooks.ir
Alopecia
97
Fig. 2 Focal alopecia on
the front limb in a kitten
affected by
dermatophytosis
Fig. 3 Multifocal alopecia
in a kitten affected by
dermatophytosis
Fig. 4 Spontaneous
alopecia following an
adverse reaction to flea
collar in an adult cat
Self-induced alopecia is caused by the cat itself by excessive licking and, less
commonly, by chewing, plucking hair, or scratching (Fig. 5). Licking in the feline
species is a major component of grooming, a normal, genetically programmed feline
behavior. Cats groom to remove dead hair, ectoparasites and dirt and to control
body temperature. One study suggested that a healthy cat grooms for approximately
1 hour per day [3]. An increased frequency and/or intensity of this behavior is called
98
VetBooks.ir
Table 1 Selected causes of
spontaneous alopecia
S. Colombo
Inflammation/infection
of the hair follicle
Dysplasia/atrophy of
the hair follicle
Fig. 5 Self-induced
alopecia in an allergic cat
Pyoderma
Dermatophytosis
Demodicosis (Demodex cati)
Pemphigus foliaceus
Pseudopelade
Lymphocytic mural folliculitis
Sebaceous adenitis
Topical/injectable glucocorticoid
administration
Topical/systemic adverse drug reaction
Telogen effluvium
Spontaneous/iatrogenic
hyperadrenocorticism
Paraneoplastic alopecia
Post-traumatic alopecia
Alopecic breeds/congenital
hypotrichoses
Pili torti
Cicatricial alopecia (scar)
Alopecia
Pruritus
VetBooks.ir
Table 2 Selected causes of
self-induced alopecia
99
Pain/neurologic
Behavioral
Pyoderma
Dermatophytosis
Malassezia overgrowth
Flea infestation
Cheyletiellosis
Otodectic mange (erratic)
Demodicosis (Demodex gatoi)
Lynxacarus infestation
Flea-bite hypersensitivity
Adverse reaction to food
Feline atopic syndrome
Allergic contact dermatitis
Feline lymphocytosis
Feline hyperesthesia syndrome
Irritant contact dermatitis
Feline idiopathic cystitis
Trauma
Psychogenic alopecia
overgrooming and may be the expression of pruritus, pain or behavioral problems
(Table 2). Being the increased expression of a physiological behavior, overgrooming is often not recognized by the owner or not interpreted as a sign of pruritus or
pain. Moreover, cats tend to express their discomfort by hiding away from the owners, who may not be aware of their pet’s overgrooming.
Finally, one must remember that some diseases may cause both spontaneous
alopecia due to damage to the hair follicle and self-induced alopecia due to pruritus.
For example, some cases of dermatophytosis or demodicosis may be associated
with pruritus.
Diagnostic Approach
Signalment and History
Infectious and ectoparasitic diseases such as dermatophytosis, demodicosis, flea
infestation, or cheyletiellosis are commonly observed in kittens or in environmental
conditions of crowding, such as breeding colonies or pet shops. Paraneoplastic syndromes and neoplasia are typically seen in older cats. Breed may be a relevant information in the diagnostic approach: Persian cats are predisposed to dermatophytosis
(Fig. 6); congenital hypotrichosis has been recently reported in Birman cats [1]. A
good knowledge of feline phenotypes is important, especially for breeds such as the
Devon rex cat, which may have an extremely variable amount of hair on the trunk
and is physiologically alopecic on the lateral and ventral neck.
History is also very relevant for the diagnosis. A scar may be easily diagnosed
based on history, while previous trauma from a car accident or a fall may point
towards a post-traumatic alopecia [4]. A detailed pharmacological history is
VetBooks.ir
100
S. Colombo
Fig. 6 Alopecia and
scaling on the tail of a
Persian cat with
dermatophytosis
Fig. 7 Spontaneous
alopecia in an old cat
affected by
hyperadrenocorticism and
demodicosis
important when an adverse drug reaction is suspected. Sudden onset of alopecia
in a queen that recently gave birth may suggest telogen effluvium. Seasonality of
self-induced alopecia may orientate towards feline atopic dermatitis. Concurrent
systemic clinical signs such as polyuria and polydipsia in an old, diabetic cat
developing spontaneous alopecia should prompt testing for hyperadrenocorticism
(Fig. 7), while self-­induced alopecia of the abdomen and groin may be caused by
feline idiopathic cystitis [5].
Clinical Presentation
Spontaneous alopecia may be partial or complete and, in general, hair can be easily epilated from the whole alopecic area, from the center of the lesion or from its
periphery. The skin looks glabrous and smooth, and few short fragments of hair can
be seen emerging from follicular ostia in selected diseases such as dermatophytosis.
VetBooks.ir
Alopecia
101
Fig. 8 Close-up image of
the abdominal skin of a cat
with self-induced alopecia
Self-induced alopecia is characterized by the presence of very short fragments of
hair which can be observed by looking closely at the skin or with the help of a magnifying lens (Fig. 8). Hair cannot be easily epilated. Self-induced alopecia is often
complete and may be symmetrical. The alopecic area usually has very well-­defined
margins, with abrupt change to normal hair.
Both spontaneous and self-induced alopecia in cats may be focal, multifocal or
generalized, and may be associated with other skin lesions. The presence/absence
and type of lesion accompanying alopecia is extremely useful to orient the diagnostic process (Table 3).
Focal alopecia and thickening of the affected skin, together with history of previous trauma, may allow the clinician to identify a scar. The skin may also be hypo- or
hyperpigmented. Mild erythema and exfoliation associated with focal or multifocal
alopecia in cats may suggest dermatophytosis. Pruritus may vary from absent to
moderate and for this reason dermatophytosis should also be considered in the list
of differential diagnoses of self-induced alopecia. A focal area of non-inflammatory
alopecia with very thin skin, visible blood vessels and bruising suggests a reaction
to one or repeated glucocorticoid injections in that site (Fig. 9). Generalized, predominantly ventral alopecia with shiny skin in an old cat is suggestive of paraneoplastic alopecia (Fig. 10) [6].
Focal or multifocal alopecia and erythema, mild scaling and occasionally comedones, associated with mild or no pruritus may be indicative of demodicosis due
to Demodex cati, a follicular mite that usually causes disease in immunocompromised animals. Severe pruritus and self-induced alopecia with erythema and scaling
raises the suspicion of demodicosis due to Demodex gatoi, a contagious, short-bodied mite living in the stratum corneum. Demodicosis is uncommon in the cat [7].
Severe scaling and self-induced alopecia, often with a dorsal distribution, may indicate cheyletiellosis. Self-induced alopecia, particularly if miliary dermatitis and/
or eosinophilic plaques are concurrently observed, may be very suggestive of an
allergic disease.
102
S. Colombo
VetBooks.ir
Table 3 Examples of lesions observed concurrently to alopecia in feline skin diseases
Spontaneous
alopecia
Self-induced
alopecia
Lesions
Erythema, scaling, follicular casts
Disease
Dermatophytosis
Erythema, scaling, comedones, follicular
casts
Papules, crusting, scaling
Pustules, yellow crusting
Onychomadesis, onychorrhexis
Scaling, hyperpigmentation
Scaling, crusting, follicular casts
Focal thinning, visible blood vessels,
bruising
None
Thin skin, bruising, tears, scaling,
comedones
Shiny skin
Erythema, shiny skin, erosions/ulcers
Absence of whiskers, claws, tongue
papillae
Thickening, hypo-/hyperpigmentation
Scaling
Demodicosis
Ceruminous otitis externa
Miliary dermatitis, eosinophilic plaque
Papules, crusting, scaling
Erythema, scaling, ceruminous otitis
externa, paronychia, chin acne
Erythema, erosions/ulcers, plaques
Skin rolling
Erosions/ulcers
None
None
Otodectic mange (erratic)
Allergic diseases
Superficial pyoderma
Malassezia overgrowth
Fig. 9 Spontaneous, focal
alopecia in a cat treated
with repeated injections of
a glucocorticoid
Superficial pyoderma
Pemphigus foliaceus
Pseudopelade
Lymphocytic mural folliculitis
Sebaceous adenitis
Topical/systemic glucocorticoid
administration
Telogen effluvium
Spontaneous/iatrogenic
hyperadrenocorticism
Paraneoplastic alopecia
Post-traumatic alopecia
Congenital hypotrichoses
Scar
Cheyletiellosis
Feline lymphocytosis
Feline hyperesthesia syndrome
Trauma
Feline idiopathic cystitis
Psychogenic alopecia
VetBooks.ir
Alopecia
103
Fig. 10 Diffuse alopecia
and shiny skin on the
abdomen of a cat with
paraneoplastic alopecia
Together with the correct distinction between spontaneous and self-induced alopecia and identification of concurrent lesions, preferential localization of the clinical signs may help in listing the differential diagnoses (Table 4).
Diagnostic Algorithm
This section is illustrated in Fig. 11. Red squares with numbers represent the steps
of the diagnostic process, as explained below.
1
Perform microscopic hair examination
Microscopic examination of hair shafts is the first test to perform in any case
of alopecia in cats since it may yield useful information, beyond the distinction
between spontaneous and self-induced alopecia.
First, the hair tips must be evaluated: broken tips indicate self-induced alopecia, while intact tips may indicate spontaneous hair loss, with the exception of
dermatophytosis. Second, the hair shaft in its entire length should be carefully
observed. Congenital abnormalities, such as pili torti, present with flattened
hair shafts that twist on their own axis by 180 degrees at irregular intervals
104
S. Colombo
VetBooks.ir
Table 4 Common locations of alopecia in selected feline skin diseases
Spontaneous alopecia
Distribution
Head, pinnae, paws, tail, generalized
Head, neck, ear canal, generalized
Head, pinnae, claw folds, abdomen
Head, abdomen, legs, paws
Head, pinnae neck, generalized
Site of application/injection
Trunk
Abdomen, ventral trunk, medial legs
Rump
Generalized
Site of previous trauma
Self-induced alopecia
Rump
Dorsum
Neck, rump, tail, ear canal
Thorax, abdomen
Rump
Abdomen, medial thighs, head, neck
Chin, claw folds, face, ear canal,
generalized
Thorax, legs, pinnae, neck
Dorsum
Abdomen, groin
Site of previous trauma
2
Disease
Dermatophytosis
Demodicosis
Pemphigus foliaceus
Pseudopelade
Sebaceous adenitis
Topical/systemic glucocorticoid administration
Spontaneous/iatrogenic hyperadrenocorticism
Paraneoplastic alopecia
Post-traumatic alopecia
Alopecic breeds/congenital hypotrichoses
Cicatricial alopecia (scar)
Flea infestation
Cheyletiellosis
Otodectic mange (erratic)
Demodicosis
Flea-bite hypersensitivity
Other allergic diseases
Malassezia overgrowth
Feline lymphocytosis
Feline hyperesthesia syndrome
Feline idiopathic cystitis
Post-traumatic alopecia
(Fig. 12) [8]. Moving towards the root, spores of dermatophytes arranged
around the hair shaft or Demodex mites free or embedded in keratin casts may
be identified. However, a negative result of hair examination does not rule out
dermatophytosis and demodicosis.
Rule out non-dermatological causes of self-induced alopecia
When the microscopic examination of hair shafts indicates self-induced alopecia, non-dermatological causes must be carefully considered and history
should specifically investigate for concurrent non-dermatological signs. If the
alopecia occurs on the abdomen and groin only, urinalysis and bacterial culture
and sensitivity testing should be performed to investigate feline idiopathic cystitis, ­urolithiasis and/or lower urinary tract infections. Ultrasound examination
may help investigate other causes of abdominal pain. If the alopecia is focal and
located, for example, on a single limb or on the dorsal spine, an x-ray examination may identify a previous trauma which may explain the cat’s continuous
licking at that site. When abnormal behavior such as rippling or rolling the skin
along the lumbar spine is reported by the owner to occur frequently, a neurological examination should be recommended [9].
Alopecia
105
Alopecia
VetBooks.ir
1
Self-induced
Pili torti
Demodicosis
Dermatophytosis
Demodicosis
Skin scraping
Dermatophytosis
Wood’s lamp examination
5
Cytology
Trauma
3
Telogen effluvium
Complete blood count
Biochemistry
Urinalysis
6
Ultrasound
Endocrinology testing
TC/MRI
Fungal culture
Feline idiopathic cystitis
Scar
Topical/systemic
glucocorticoid administration
4
X-ray
Urinalysis
Bacterial culture
and sensitivity testing
Ultrasound
Neurologic examination
(if appropriate)
Feline hyperesthesia syndrome
Spontaneous
History
Superficial pyoderma
2
Hair examination
Go to chapter 9
Pruritus
Spontaneous/iatrogenic
hyperadrenocorticism
7
Pemphigus foliaceus
Histopathology
Paraneoplastic alopecia
8
X-ray
Post-traumatic alopecia
Pseudopelade
Lymphocytic mural folliculitis
Sebaceous adenitis
Congenital hypotrichoses
Topical/systemic adverse drug reaction
Fig. 11 Diagnostic algorithm of alopecia
Fig. 12 Microscopic
examination of the hair
shaft of a cat with pili torti
(10X)
3
Finally, if all these potential causes of alopecia do not comply with the history and clinical presentation or have been ruled out, self-induced alopecia
should be further investigated following the diagnostic approach to pruritus
(Chapter, Pruritus).
Consider the patient’s history
Spontaneous focal alopecia associated with variations of skin thickness and
history of a wound in that site points towards a diagnosis of scar. If a topical
glucocorticoid has been applied or a glucocorticoid injection has been given
VetBooks.ir
106
4
5
6
7
8
S. Colombo
where alopecia developed and the skin appears thin, with bruising and visible
vessels, the diagnosis is straightforward and may be supported by the observation of mostly telogen hair roots on microscopic examination. Sudden onset of
diffuse alopecia in a queen who recently gave birth, for example, may suggest
telogen effluvium. In this case, the remaining hair is easily epilated and microscopic examination of hair shows telogen roots only.
Perform skin scrapings
Skin scrapings are diagnostic for demodicosis and, together with microscopic examination of hair, may be strongly suggestive of dermatophytosis. In
fact, the correct identification of dermatophyte spores surrounding hair shafts
may be easier on skin scrapings than microscopic examination of hair shafts,
because scraping the surface of the alopecic area is likely to collect more broken, infected hair [10].
Perform Wood’s lamp examination and fungal culture
These two diagnostic tests, taken together, are diagnostic for dermatophytosis or, if negative results are obtained, are helpful to rule it out. Since dermatophytosis is the most common cause of alopecia in cats, a fungal culture is
appropriate in all cases presenting with alopecia.
Consider non-dermatological clinical signs
In an old cat presenting with alopecia and systemic signs such as polyuria/
polydipsia, polyphagia, vomiting or weight loss, one must consider the possibility of alopecia being caused by a systemic disease. If the alopecic skin appears
thin, with bruising and/or tears developing after minimal traction, the cat should
be investigated for hyperadrenocorticism. History may suggest iatrogenic
hyperadrenocorticism, if glucocorticoids have been administered for a long
time, or spontaneous hyperadrenocorticism if there is no history of glucocorticoids administration or the cat is diabetic. Ventrally distributed alopecia with
shiny skin in a cat presenting with concurrent weight loss, depression, vomiting,
­and/or diarrhea may point towards a diagnosis of paraneoplastic alopecia and
should prompt to perform an abdominal ultrasound examination.
Perform cytology
Cytology should be performed if other lesions, such as pustules, crusts or
erosions/ulcers are present together with alopecia. The observation of large
numbers of degenerate neutrophils with intracellular and extracellular bacteria
indicates superficial pyoderma. If the neutrophils appear “healthy” and many
acantholytic keratinocytes are seen, the results of cytological examination are
suggestive of pemphigus foliaceus. If large numbers of eosinophils are seen, it
is more likely that the cat is pruritic and alopecia should be further investigated
following the diagnostic approach to pruritus (Chapter, Pruritus).
Take biopsies for histopathological examination
Histopathological examination may be useful to confirm paraneoplastic alopecia and should always be performed if pemphigus foliaceus is suspected.
Some diseases presenting with alopecia can only be diagnosed with histopathology;
examples are pseudopelade, sebaceous adenitis, congenital hypotrichosis, and
VetBooks.ir
Alopecia
107
adverse drug reactions. If the histopathological examination is suggestive of
post-traumatic alopecia, radiological examination of the pelvis should be carried out to confirm the diagnosis.
References
1. Abitbol M, Bossé P, Thomas A, Tiret L. A deletion in FOXN1 is associated with a syndrome
characterized by congenital hypotrichosis and short life expectancy in Birman cats. PLoS One.
2015;10:1–12.
2. Genovese DW, Johnson TL, Lamb KE, Gram WD. Histological and dermatoscopic description
of sphynx cat skin. Vet Dermatol. 2014;25:523–e90.
3. Eckstein RA, Hart BL. The organization and control of grooming in cats. Appl Anim Behav
Sci. 2000;68:131–40.
4. Declerq J. Alopecia and dermatopathy of the lower back following pelvic fractures in three
cats. Vet Dermatol. 2004;15:42–6.
5. Amat M, Camps T, Manteca X. Stress in owned cats: behavioural changes and welfare implications. J Feline Med Surg. 2016;18:1–10.
6. Turek MM. Cutaneous paraneoplastic syndromes in dogs and cats: a review of the literature.
Vet Dermatol. 2003;14:279–96.
7. Beale K. Feline Demodicosis. A consideration in the itchy or overgrooming cat. J Feline Med
Surg. 2012;14:209–13.
8. Maina E, Colombo S, Abramo F, Pasquinelli G. A case of pili torti in a young adult domestic
short-haired cat. Vet Dermatol. 2013;24:289–e68.
9. Ciribassi J. Feline hyperesthesia syndrome. Compend Contin Educ Vet. 2009;31:116–22.
10. Colombo S, Cornegliani L, Beccati M, Albanese F. Comparison of two sampling methods for
microscopic examination of hair shafts in feline and canine dermatophytosis. Vet Dermatol.
2008;19(Suppl. 1):36.
General References
For definitions: Merriam-Webster Medical Dictionary. http://merriam-webster.com Accessed 10
May 2018.
Albanese F. Canine and feline skin cytology. Cham: Springer International Publishing; 2017.
Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, Wolff K. Fitzpatrick’s dermatology in
general medicine. 8th ed. New York: The McGraw-Hill Companies; 2012.
Mecklenburg L. An overview on congenital alopecia in domestic animals. Vet Dermatol.
2006;17:393–410.
Miller WH, Griffin CE, Campbell KL. Muller & Kirk’s small animal dermatology. 7th ed. St.
Louis: Elsevier; 2013.
Noli C, Toma S. Dermatologia del cane e del gatto. 2nd ed. Vermezzo: Poletto Editore; 2011.
VetBooks.ir
Papules, Pustules, Furuncles and Crusts
Silvia Colombo
Abstract
Papules, pustules, furuncles, abscesses and crusts are common lesions in cats.
With the exception of abscesses, they are often observed in combinations, representing different stages of the same disease evolving into one another. In general,
these lesions are the expression of inflammatory diseases, with infectious, parasitic, allergic or autoimmune pathogenesis. Clinical presentations of papules,
pustules, furuncles, abscesses and crusts and their preferential localization in
selected feline diseases are described, together with useful diagnostic hints coming from signalment and history. A feline-specific clinical presentation called
miliary dermatitis is characterized by multiple, small crusted papules and pruritus. The diagnostic approach to papules, pustules, furuncles, abscesses and crusts
requires performing the diagnostic tests in a systematic way. Dermatophytosis is
very common in cats, and diagnostic tests to diagnose or rule out this disease
should be carried out in all cases presenting with papules, pustules, crusts or as
miliary dermatitis.
Definitions
A papule is a solid, erythematous, elevated skin lesion of less than 1 cm diameter [1].
Many papules close to each other may coalesce to form a plaque (Chapter, Plaques,
Nodules and Eosinophilic Granuloma Complex Lesions).
A pustule is an elevated, circumscribed, hollow lesion containing pus and covered by epidermis. It may be centered around a hair follicle or may be interfollicular
S. Colombo (*)
Servizi Dermatologici Veterinari, Legnano, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_5
109
VetBooks.ir
110
S. Colombo
in location. Pustules usually contain neutrophils, with or without bacteria, or less
commonly eosinophils. They are fragile and often transient lesions, uncommonly
observed in cats.
A furuncle is similar to a pustule, but it is larger in size and deeper in location,
because it results from the complete destruction of the hair follicle. The wall of a
furuncle is thicker than the roof of a pustule, and its content comprises pus, blood
(in this case, it is also called hemorrhagic bulla) or a mixture. It is usually a very
inflamed and painful lesion, centered around a hair follicle. The furuncle may open
and drain pus, blood or a hemopurulent exudate.
An abscess is a circumscribed, fluctuant, dermal or subcutaneous collection of
pus. It may open and drain on the skin surface, forming a draining tract.
A crust is an accumulation of dried exudate. The crust is yellowish when the
dried material is pus, or brownish if dried blood is its main component (hemorrhagic
crust). It may also contain microorganisms and epidermal cells, such as acantholytic
keratinocytes or corneocytes and, if the crust encloses a tuft of hair, its removal
results in focal alopecia.
Pathogenesis
Papules, pustules and crusts represent collections of inflammatory cells in the epidermis (pustule), dermis (papule) or on the skin surface (crust) as dead remnants
of these cells. The inflammatory cells are attracted towards the superficial layers of
the skin by infectious agents, parasites or allergens, or may be the expression of an
autoimmune disease such as pemphigus foliaceus (Fig. 1).
The furuncle is a deeper lesion which results from the complete destruction of the
hair follicle. The hair follicle is destroyed by severe inflammation, which is, in the
feline species, most commonly induced by a bacterial infection as in complicated
Fig. 1 Severe crusting due
to drying of purulent
exudate on the pinna of a
cat with pemphigus
foliaceus
VetBooks.ir
Papules, Pustules, Furuncles and Crusts
111
Fig. 2 Furuncles on the
chin of a cat affected by
complicated chin acne
chin acne (Fig. 2) [2]. The hair shaft may be free in the dermis together with bacteria
and other debris, and attracts more inflammatory cells behaving as a foreign body.
The abscess usually occurs following bite or claw wounds, with implantation
of bacteria in the deep dermis and subcutis. The presence of bacteria attracts large
numbers of neutrophils and other inflammatory cells at the infection site, until a
large collection of pus is formed (Fig. 3).
Papules, pustules, furuncles and crusts may represent different stages of the same
disease and can evolve into one another. A papule may develop into a pustule, which
ruptures and becomes a small crust. Very uncommonly in the cat, a circular rim
of scales may form when the crust comes off: this lesion is called epidermal collarette. A pustule may become a furuncle if the infection deepens and extends to
involve and destroy the whole hair follicle. If the furuncle opens and drains exudate,
a crust may form. When the crust comes off, an area of focal alopecia is the final
result. Crusts may also cover other lesions, such as erosions and ulcers (Chapter,
Excoriations, Erosions and Ulcers) (Fig. 4). This is important to keep in mind when
examining the animal, because we may be able to identify different lesions which
represent evolving stages of the disease or we may only find the final result of
112
S. Colombo
VetBooks.ir
Fig. 3 Retroauricular
abscess in a stray cat
Fig. 4 Hemorrhagic crust
covering an erosion/ulcer
on the nose of a cat with
herpesvirus infection
this process, which is the crust. Table 1 lists selected causes of papules, pustules,
abscesses, crusts and furuncles in cats.
Diagnostic Approach
Signalment and History
Contagious diseases such as notoedric mange and dermatophytosis are most commonly observed in kittens, while neoplasia is typically seen in older cats. Cutaneous
abscesses occur more often in intact male cats, as a consequence of fighting. Breed
may be a relevant point in the diagnostic approach: Persian cats are predisposed
VetBooks.ir
Papules, Pustules, Furuncles and Crusts
Table 1 Selected causes of
papules, pustules, furuncles,
abscesses and crusts
Papules
Pustules
Furuncles
Abscess
Crusts
113
Notoedric mange
Dermatophytosis
Mosquito-bite hypersensitivity
Allergic diseases
Urticaria pigmentosa-like dermatitis
Xanthomas
Mast cell tumor
Pemphigus foliaceus
Complicated chin acne
Bacterial infections
Trauma (including self-inflicted)
Pyoderma
Notoedric mange
Dermatophytosis
Subcutaneous and systemic fungal
infections
Herpesvirus dermatitis
Poxvirus infection
Allergic diseases
Mosquito-bite hypersensitivity
Adverse drug reactions
Pemphigus foliaceus
Complicated chin acne
Perforating dermatitis
Idiopathic facial dermatitis of Persian and
Himalayan cats
Squamous cell carcinoma
Idiopathic/behavioral ulcerative dermatitis
to dermatophytosis and idiopathic facial dermatitis [3]. Urticaria pigmentosa-­like
dermatitis has been described in Devon rex and Sphynx cats [1, 4].
History is obviously of paramount importance for the diagnosis when previous trauma (including self-induced) is suspected in a cat examined for a crusting
lesion. Especially in kittens, detailed information on where the pet was acquired
must always be collected. Being found as a stray or adopted from a cattery may
represent a predisposing factor for dermatophytosis, notoedric mange and herpesvirus dermatitis. Lifestyle is also relevant, because outdoor cats may be affected by
mosquito-­bite hypersensitivity and development of abscesses more commonly than
indoor cats. Regularly hunting mice and voles is a predisposing factor for Poxvirus
infection. Contagion of in-contact pets or people should prompt investigation for
dermatophytes and ectoparasites.
Last but not least, one very important question to ask when taking the history is
whether the cat is pruritic or not, and if pruritus is continuously present or seems to
occur at a specific time of the year. Notoedric mange is a severely pruritic disease,
and seasonal pruritus may suggest flea-bite or mosquito-bite hypersensitivity, and
feline atopic syndrome.
114
S. Colombo
VetBooks.ir
Clinical Presentation
Papules and pustules are in most cases multiple lesions, sometimes with a grouped
configuration. In feline urticaria pigmentosa-like dermatitis, papules may have a
linear configuration [1]. A single or many furuncles may be observed in chin acne.
The distribution of papules, pustules, furuncles and crusts may be localized or generalized. The abscess is usually a single lesion.
Papules and pustules are primary skin lesions; however, in most diseases, they
represent one step in a pathological continuum of lesions. For example, although
papules are the primary lesions in notoedric mange, they may not be visible, because
they are covered by very thick crusts. Multiple, erythematous small papules covered by crusts, especially on the dorsum, may develop representing a feline-specific
clinical presentation called miliary dermatitis [5, 6] (see later) (Fig. 5). The location
of the lesions on the cat’s body may be helpful in developing a correct list of differential diagnoses (Table 2).
An erythematous to hyperpigmented papular eruption, which may have a linear
distribution on the ventrolateral chest and abdomen, is often pruritic and occurs in
a Devon rex or Sphynx cats, is consistent with urticaria pigmentosa-like dermatitis
(Fig. 6) [1, 4]. Small, erythematous, and crusted papules may suggest mosquito-bite
hypersensitivity, when distributed on the dorsal nose, pinnae and footpads (Fig. 7)
[7]. Pustules may be difficult to observe because they are transient, fragile lesions,
but, when observed on the face, inner pinnae and abdomen, close to the nipples and
on footpads should prompt investigation for pemphigus foliaceus (Fig. 8) [8].
Furuncles in cats are usually observed on the chin, where they develop when
chin acne becomes complicated by secondary bacterial infection. A soft, fluctuant
swelling, occasionally with a draining tract from which purulent exudate comes out,
located on the face, neck, or tail base, most likely represents an abscess.
Crusts are extremely common lesions, as they are the final result of the pathological continuum of lesions described in this chapter, as well as of traumatic lesions.
Fig. 5 Miliary dermatitis
in an allergic cat
VetBooks.ir
Papules, Pustules, Furuncles and Crusts
Table 2 Common locations
of papules, pustules,
abscesses, crusts and
furuncles in selected feline
skin diseases
Papules
Distribution
Head, pinnae, neck,
paws, perineum
Head, pinnae, paws, tail,
generalized
Head, pinnae, paws
Rump
Head, paws, bony
prominence
Pustules
Head, pinnae, claw
folds, abdomen
Furuncles
Chin
Abscesses
Neck, shoulders, tail
base
Crusts
Site of previous trauma
Face
Face, ear canals
Head, pinnae
115
Disease
Notoedric mange
Dermatophytosis
Mosquito-bite
hypersensitivity
Flea-bite hypersensitivity
Xanthomas
Pemphigus foliaceus
Complicated chin acne
Bacterial infections
Trauma
Herpesvirus dermatitis
Idiopathic facial dermatitis
of Persian and Himalayan
cats
Squamous cell carcinoma
Fig. 6 Multiple
erythematous papules in a
Devon rex cat with
urticaria pigmentosa-like
dermatitis
One helpful clinical hint, when crusts are observed, is their color. If the crusts
are dark brown, they are composed by dried blood and the lesion was most likely
caused by a deep skin disease (ulcer) or (self-)trauma. If they are yellow, they represent dried purulent material and intact pustules should be carefully searched for.
Very thick and dry light-colored crusts on the head, margins of pinnae, neck, paws,
VetBooks.ir
116
S. Colombo
Fig. 7 Papules on the
pinna of a cat affected by
mosquito-bite
hypersensitivity
and perineum, associated with severe pruritus, are the predominantly observed
lesions in notoedric mange. Multiple, conical, very dry and thick crusted lesions
(eschars) developing at sites of previous trauma may indicate a rare feline disease
called acquired reactive perforating collagenosis or perforating dermatitis (Fig. 9)
[9]. These lesions are difficult to remove and usually cover an ulcerated, hemorrhagic area. Pruritus and adherent, black, variably dried exudate covering areas of
erythema or erosions distributed around the eyes, mouth, and chin are typical of
idiopathic facial dermatitis of Persian and Himalayan cats, also called dirty face
disease [3].
VetBooks.ir
Papules, Pustules, Furuncles and Crusts
117
Fig. 8 Pustules and
crusting on the inner pinna
of a cat with pemphigus
foliaceus
Miliary Dermatitis
Miliary dermatitis is a peculiar clinical presentation observed only in the cat. It is
characterized by small, crusted papules “resembling millet seeds”, hence the name,
which are more easily felt by touching through the haircoat then seen. Miliary dermatitis mainly affects the trunk and neck and is often associated with pruritus and selfinduced alopecia (Fig. 10) [5, 6]. Differential diagnoses of miliary dermatitis are listed
in Table 3. Miliary dermatitis should be investigated following the diagnostic approach
to pruritus (Chapter, Pruritus).
VetBooks.ir
118
S. Colombo
Fig. 9 Dry, thick, adherent
yellow crust on the inner
pinna of a young cat with
perforating dermatitis
Fig. 10 Alopecia and
miliary dermatitis on the
dorsum of a cat affected by
flea-bite hypersensitivity
Table 3 Differential
diagnoses of miliary
dermatitis
Miliary dermatitis
Cheyletiellosis
Other ectoparasites (Lynxacarus
radowski)
Dermatophytosis
Flea-bite hypersensitivity
Adverse reaction to food
Feline atopic syndrome
Adverse drug reaction
Pemphigus foliaceus
Papules, Pustules, Furuncles and Crusts
119
VetBooks.ir
Diagnostic Algorithm
This section is illustrated in Fig. 11. Red squares with numbers represent the steps
of the diagnostic process, explained below.
1
2
3
4
Consider signalment, history and physical examination
Signalment, history, and physical examination may give the clinician
extremely useful information for the diagnostic process. In a primarily outdoor
intact male cat presenting with a fluctuant mass on the neck, for example, the
most likely diagnosis is an abscess. When the main presenting signs are furuncles on the chin of a cat that suffers from chin acne, it is very likely that acne has
become secondarily complicated by a bacterial infection. If physical examination reveals papules, pustules or crusts, a standardized sequence of diagnostic
tests is usually required to make the diagnosis.
Perform skin scrapings
Skin scrapings must be performed whenever papules, pustules, crusts or
furuncles are observed. Skin scrapings are diagnostic for notoedric mange and
may identify Demodex cati mites in cases of chin furunculosis [10].
Perform Wood’s lamp examination and fungal culture
These two diagnostic tests, taken together, are diagnostic for dermatophytosis
or, if negative results are obtained, are helpful to rule it out. Since dermatophytosis may present with papules, pustules, miliary dermatitis and crusts in cats, a
fungal culture is appropriate in all cases presenting with these lesions (Fig. 12).
Perform cytology
When the physical examination reveals the presence of an abscess, cytology
from the purulent exudate should always be performed to support the diagnostic
hypothesis. Usually, large numbers of degenerate neutrophils are visible,
admixed with bacteria and variable numbers of macrophages, lymphocytes and
plasma cells. To identify the bacteria species causing the abscess, bacterial culture and sensitivity testing should be performed. It is also advisable testing cats
with abscesses for FIV and FeLV. Cytological examination of exudate draining
from furuncles on the chin usually shows pyogranulomatous inflammation with
bacteria. Bacterial culture and sensitivity testing for aerobes and anaerobes may
be required to identify the causative microorganism and choosing the most
effective antibiotic for treatment, if needed [2].
Papules, pustules and crusts should always be investigated by cytological
examination, a simple test that often gives very useful information. The observation of large numbers of non-degenerate neutrophils admixed with many acantholytic keratinocytes suggests pemphigus foliaceus. Eosinophilic inflammation
is very common in cats. If eosinophils are present in large numbers within a
mixed inflammatory infiltrate in samples obtained from crusted, papular lesions
on the bridge of the nose, mosquito-bite hypersensitivity is a likely diagnosis [7].
Skin scraping
6
Histopathology
Miliary dermatitis
History
Pruritus
Physical examination
Papules
Pustules
Crusts
5
Signalment, History, Physical examination
Fig. 11 Diagnostic algorithm to papules, pustules, furuncles, abscesses and crusts
Herpesvirus dermatitis
Poxvirus infection
Subcutaneous and systemic fungal infections
Adverse drug reactions
Perforating dermatitis
Idiopathic facial dermatitis of Persian and Himalayan cats
4
Cytology
Fungal culture
3
Wood’s lamp examination
Subcutaneous and systemic fungal
infections
Pemphigus foliaceus
Mosquito-bite hypersensitivity
Urticaria pigmentosa-like dermatitis
Xanthomas
Mast cell tumor
Squamous cell carcinoma
Dermatophytosis
Notoedric mange
2
1
Skin scraping
4
Complicated chin acne
Cytology
Bacterial culture and
sensitivity testing
FIV-FeLV serology
Go to chapter 9
Pruritus
Chin acne and demodicosis
2
Furuncles
Abscess
Papules, Pustules, Furuncles, Abscess, Crusts
VetBooks.ir
120
S. Colombo
VetBooks.ir
Papules, Pustules, Furuncles and Crusts
5
121
Neutrophils, eosinophils and occasionally mast cells observed in samples from
erythematous, hyperpigmented papules on the skin of a Devon rex or Sphynx
cat are suggestive of urticaria pigmentosa-like dermatitis [4]. Finally, cytological examination may show a monomorphic population of well-differentiated
mast cells in mast cell tumors or epithelial cells in small aggregates or as single
cells, with aspects of squamous differentiation, often admixed with neutrophils
and other inflammatory cells, in squamous cell carcinomas (Fig. 13). Cytological
findings obtained from crusting, papular or pustular lesions must always be confirmed by biopsy and histopathological examination.
Consider history, pruritus and physical examination
When skin scrapings, Wood’s lamp examination and fungal culture yield
negative results and cytological findings are nonspecific (e.g., neutrophilic
inflammation), one must carefully re-consider the history and clinical findings.
In a continuously or seasonally pruritic cat, presenting with crusted papules on
Fig. 12 Alopecia and
crusting on the face of a
cat with dermatophytosis
Fig. 13 Hemorrhagic
crusting on the nose of a
cat with squamous cell
carcinoma
VetBooks.ir
122
6
S. Colombo
the dorsum, or, less commonly, with generalized distribution, miliary dermatitis
should be further investigated following the diagnostic approach to pruritus
(Chapter, Pruritus).
Take biopsies for histopathological examination
Histopathological examination should always be performed if pemphigus
foliaceus, mosquito-bite hypersensitivity, urticaria pigmentosa-like dermatitis,
or infectious, metabolic and neoplastic diseases are suspected, based on cytological findings. Other diseases with nonspecific cytological findings and
requiring histopathological examination for the diagnosis are, for example, viral
diseases, perforating dermatitis, idiopathic facial dermatitis of Persian and
Himalayan cats and adverse drug reactions.
References
1. Vitale C, Ihrke PJ, Olivry T, Stannard AA. Feline urticaria pigmentosa in three related Sphinx
cats. Vet Dermatol. 1996;7:227–33.
2. Jazic E, Coyner KS, Loeffler DG, Lewis TP. An evaluation of the clinical, cytological, infectious and histopathological features of feline acne. Vet Dermatol. 2006;17:134–40.
3. Bond R, Curtis CF, Ferguson EA, Mason IS, Rest J. An idiopathic facial dermatitis of Persian
cats. Vet Dermatol. 2000;11:35–41.
4. Noli C, Colombo S, Abramo F, Scarampella F. Papular eosinophilic/mastocytic dermatitis
(feline urticaria pigmentosa) in Devon rex cats: a distinct disease entity or a histopathological
reaction pattern? Vet Dermatol. 2004;15:253–9.
5. Hobi S, Linek M, Marignac G, Olivry T, Beco L, Nett C, et al. Clinical characteristics and
causes of pruritus in cats: a multicentre study on feline hypersensitivity-associated dermatoses.
Vet Dermatol. 2011;22:406–13.
6. Diesel A. Cutaneous hypersensitivity dermatoses in the feline patient: a review of allergic skin
disease in cats. Vet Sci. 2017:25. https://doi.org/10.3390/vetsci4020025.
7. Nagata M, Ishida T. Cutaneous reactivity to mosquito bites and its antigens in cats. Vet
Dermatol. 1997;8:19–26.
8. Olivry T. A review of autoimmune skin diseases in domestic animals: I – superficial pemphigus. Vet Dermatol. 2006;17:291–305.
9. Albanese F, Tieghi C, De Rosa L, Colombo S, Abramo F. Feline perforating dermatitis resembling human reactive perforating collagenosis: clinicopathological findings and outcome in
four cases. Vet Dermatol. 2009;20:273–80.
10. Beale K. Feline demodicosis: a consideration in the itchy or overgrooming cat. J Feline Med
Surg. 2012;14:209–13.
General References
For definitions: Merriam-Webster Medical Dictionary. http://merriam-webster.com Accessed 10
May 2018.
Albanese F. Canine and feline skin cytology. Cham: Springer International Publishing; 2017.
Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, Wolff K. Fitzpatrick’s dermatology in
general medicine. 8th ed. New York: The McGraw-Hill Companies; 2012.
Miller WH, Griffin CE, Muller CKL. Kirk’s small animal dermatology. 7th ed. St. Louis: Elsevier;
2013.
Noli C, Foster A, Rosenkrantz W. Veterinary allergy. Chichester: Wiley Blackwell; 2014.
Noli C, Toma S. Dermatologia del cane e del gatto. 2nd ed. Vermezzo: Poletto Editore; 2011.
VetBooks.ir
Plaques, Nodules and Eosinophilic
Granuloma Complex Lesions
Silvia Colombo and Alessandra Fondati
Abstract
Plaques and nodules, including the lesions belonging to the eosinophilic granuloma complex (EGC), are common in cats. Plaques and nodules are caused in
most cases by infectious, allergic, metabolic or neoplastic diseases. Clinical presentations of plaques and nodules and their preferential localization in selected
feline diseases are described, together with useful hints coming from signalment
and history. A feline-specific group of plaques or nodules, known as the EGC,
and its specific features are also addressed in this chapter. EGC traditionally
comprises eosinophilic plaque (EP), eosinophilic granuloma (EG) and lip (indolent) ulcer (LU). The diagnostic approach to plaques and nodules starts with the
cytological examination, which may help the clinician to differentiate between
the neoplastic and the inflammatory nature of the lesion. Histopathological
examination is required to make or to confirm the diagnosis, and further testing
is usually suggested by the histopathological diagnosis.
Definitions
A plaque is a flat elevation of the skin greater than 1 cm of diameter, and its size is,
by definition, larger than its height. Plaques often form from a papule increasing in
size or by coalescence of multiple papules.
S. Colombo (*)
Servizi Dermatologici Veterinari, Legnano, Italy
A. Fondati
Veterinaria Trastevere - Veterinaria Cetego, Roma, RM, Italy
Clinica Veterinaria Colombo, Camaiore, LU, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_6
123
VetBooks.ir
124
S. Colombo and A. Fondati
A nodule is a solid, palpable and circumscribed skin lesion greater than 1 cm of
diameter. Nodules can be further characterized by their deepness, as epidermal,
dermal or subcutaneous nodules. Nodules may open toward the skin surface and a
draining tract may develop, with exudate of variable aspect and consistency coming
out of the lesion. A peculiar type of nodule is the cyst, which is a cavity containing
fluid or semisolid material lined by an epithelial wall.
Both nodules and plaques may be described by adding features such as number,
size, shape, color, consistency (e.g., hard or soft), surface changes (e.g., alopecic,
eroded, ulcerated) and relationship with the surrounding tissues (e.g., fixed, movable).
A soft, fluctuant, circumscribed nodule containing a collection of pus is called abscess,
and is described in Chapter, Papules, Pustules, Furuncles and Crusts. Other relevant
descriptors are whether the lesion is pruritic or non-pruritic and whether it is painful or
painless.
Plaques and nodules are common in cats and are the primary lesions of two of the
clinical presentations of the eosinophilic granuloma complex (EGC). EGC traditionally comprises eosinophilic plaque (EP), eosinophilic granuloma (EG), and lip
(indolent) ulcer (LU). These lesions affect the skin, lips and oral cavity of cats and
have been initially grouped together because they were observed simultaneously on
the same cat, therefore suggesting a common underlying cause. The EGC can be
considered a “complex” in all respects, indeed, because EP, EG, and LU share clinical and histopathological aspects and a common etiopathogenesis, in which eosinophils play a pivotal role.
Pathogenesis
A plaque is a flat, solid lesion due to infiltration of inflammatory or neoplastic cells
in the skin. In cats, it is most commonly associated with allergic or neoplastic diseases. It may develop because a papule increases in size or because many papules
coalesce. In feline dermatology, the term plaque is most often used to describe the
EP, a specific lesion belonging to the EGC (Fig. 1).
Fig. 1 Eosinophilic
plaque in a flea-allergic cat
VetBooks.ir
Plaques, Nodules and Eosinophilic Granuloma Complex Lesions
125
EGC is not a definitive diagnosis. It should be rather considered a cutaneous
reaction pattern most likely incited by underlying allergic causes, including hypersensitivity reactions to flea and, less commonly, environmental and food allergens.
Occasionally, stings or bites of arthropods other than fleas might be considered as
triggering factors for cutaneous eosinophil recruitment. However, in some cases, no
external inciting stimuli can be identified and EGC lesions remain idiopathic.
However, it must be taken into account that the reliability of current available diagnostic procedures does not always allow to definitely confirm/exclude hypersensitivity reactions towards environmental allergens in the cat.
Based on observations of EGC in family-related cats, a genetic, inheritable “dysregulation” of the eosinophil response has been suggested to predispose to the
development of EGC in the absence of detectable underlying causes, particularly in
kittens.
A combined genetic and allergic etiopathogenesis has been also suggested for
EGC [1]. A genetic predisposition to develop intense eosinophil responses might
help to explain why only a few cats develop EGC lesions, whereas the hypothesized
underlying allergic stimuli are so largely distributed and more commonly associated
with different reaction patterns, such as head and neck pruritus, self-induced alopecia or miliary dermatitis. On the other hand, a genetically based “abnormal” eosinophil response would not fit with clustering of cases in unrelated in-contact cats or
with the lack of predisposition to develop extra-cutaneous eosinophilic diseases in
cats suffering from EGC.
Nodules also develop because of infiltration of inflammatory or neoplastic cells,
however they are usually not flat and may extend deeper in the dermis and subcutaneous tissue. Non-neoplastic nodules may be induced by infectious agents, such as
bacteria or fungi, or may be sterile, as it happens in EG or sterile nodular panniculitis. Uncommon causes of nodules or, rarely, plaques in cats are foreign bodies and
deposition of calcium or lipids in the skin (Fig. 2) [2].
Cysts may be caused by congenital defects of development of different skin components or by obstruction of a sebaceous/apocrine duct (Fig. 3) [3]. Table 1 lists the
most common causes of plaques and nodules in cats.
Fig. 2 Nodule of
calcinosis cutis on the chin
of a cat with chronic
kidney disease
VetBooks.ir
126
S. Colombo and A. Fondati
Fig. 3 Multiple cysts on
the muzzle of a Persian cat
with feline cystomatosis.
(Courtesy of Dr. Stefano
Borio)
Table 1 Selected causes
of plaques and nodules
Plaques Eosinophilic plaque/granuloma
Lip ulcer
Papillomavirus infections
Xanthomas
Bowenoid in situ carcinoma
Cutaneous lymphocytosis
Mast cell tumor
Progressive feline histiocytosis
Nodules Botryomycosis
Leprosy
Rapidly growing mycobacterial infections
Nocardiosis
Dermatophytic mycetoma
Eumycotic mycetomas
Pheohyphomycosis
Sporotrichosis
Cryptococcosis
Leishmaniosis
Eosinophilic granuloma
Calcinosis cutis
Xanthomas
Sterile granuloma/pyogranuloma syndrome
Feline progressive histiocytosis
Sterile nodular panniculitis
Plasma cell pododermatitis
Squamous cell carcinoma
Basal cell tumors
Follicular tumors
Hemangiosarcoma
Lymphangiosarcoma
Mast cell tumor
Sarcoid
Vaccine-site fibrosarcoma
Epitheliotropic/non-­epitheliotropic cutaneous lymphoma
Melanocytoma/melanoma
Ceruminous cystomatosis
Plaques, Nodules and Eosinophilic Granuloma Complex Lesions
127
VetBooks.ir
Diagnostic Approach
Signalment and History
Plaques and nodules are usually observed in adult or older cats and are due to infectious, allergic, metabolic or neoplastic diseases in the majority of cases. Nodular
lesions with breed predisposition are dermatophytic mycetoma (Fig. 4) and apocrine cystomatosis, occurring more commonly in Persian cats, and mast cell tumor,
being more often diagnosed in Siamese cats [4].
History should investigate the cat’s lifestyle, since most bacterial and fungal
infections presenting with nodules require a penetrating wound to develop. For this
reason, these diseases are more likely to occur in cats allowed to go outdoors. More
specifically, contact with pigeon droppings has been advocated in cryptococcosis
and contact with decaying plant material in sporotrichosis, while leprosy syndrome
is usually reported in hunting or fighting cats [5, 6]. History of travelling or living
in endemic areas may suggest diseases such as leishmaniosis, which occurs in specific geographic locations [7].
When a cat is presented for a nodular lesion, neoplastic diseases should always
be included in the list of differential diagnoses. Useful information may be gathered
by enquiring about the age and time of lesion development, changes in its appearance and size and concurrent systemic signs presented by the cat. Vaccinal history
is also very relevant, because cats are predisposed to vaccination-site fibrosarcoma
(Fig. 5) [8]. History of prolonged sun exposure in a white cat may suggest squamous cell carcinoma (Fig. 6).
EGC lesions may be observed in cats of any breed, sex, and age; however, they
frequently occur in young cats and occasionally appear in few month-old kittens.
Lesions onset varies from acute (a few days) in EP to slow in LU and EG. Pruritus
varies from intense in EP to variable in EG and absent in LU. If pruritus is absent
and lesions are not clearly visible, as in selected cases of linear EG on caudal thighs,
Fig. 4 Large nodule on
the leg of a Persian cat
affected by dermatophytic
mycetoma
VetBooks.ir
128
S. Colombo and A. Fondati
Fig. 5 Relapse of
vaccination-site
fibrosarcoma in a cat
Fig. 6 Large nodules
diagnosed as squamous
cell carcinoma on the
dorsum of a congenitally
alopecic cat
lesions are usually identified by the owner when touching the cat (Fig. 7). Normally,
EGC lesions are chronically persistent or recurrent, but, especially in kittens, EG
may spontaneously regress with no further relapses.
Clinical Presentation
Plaques are, in most cases, lesions belonging to the EGC and may be single or more
commonly multiple. Clinical features of the EGC, including EP, EG, and LU, have
been well delineated and are considered quite distinctive [9]. The EP appears as
intensely pruritic, oozing, eroded, firm, coalescing papules and plaques affecting
sites accessible to being licked, such as ventral abdomen and inner thighs. Secondary
bacterial infection and regional lymphadenopathy are common [10].
Plaques, Nodules and Eosinophilic Granuloma Complex Lesions
129
VetBooks.ir
Fig. 7 Linear granuloma
on the hind limb of a cat
EG classically occurs as firm, yellowish, variably pruritic, alopecic, erythematous and crusting papules and plaques with a striking linear configuration when
affecting the caudal thigh. EG may also appear as single, yellowish papulo-nodular
lesions located anywhere on the body, including paws, mid-lower lip/chin, lip commissure and oral cavity. Pedal EG lesions are frequently ulcerated and crusted
whereas mucosal lesions appear as irregularly surfaced yellowish nodules, frequently located on the tongue and the palate.
LU refers to an apparently non-pruritic and non-painful, reddish-brown to yellowish, glistening, non-bleeding, well-circumscribed, frequently concave ulcer with
raised margins and the aspect of an ulcerated plaque rather than a true ulcer. The LU
occurs most commonly on the midline of the upper lip, at the philtrum or adjacent
to the upper canine tooth, mono- or bilaterally (Fig. 8).
EGC lesions with overlapping features of more than one form are commonly
observed and lesion definition can be difficult, as is the case of solitary or linearly
grouped ulcerated EG resembling LU or EP. Lesions might be therefore described
as papules, plaques and nodules belonging to the EGC, with no further clinical distinction. This observation raises the question on the adequacy of the currently
130
S. Colombo and A. Fondati
VetBooks.ir
Fig. 8 Bilateral ulcer on
the upper lips
Fig. 9 Single,
erythematous, and
exfoliative nodule on the
front limb of a cat with
epitheliotropic cutaneous
lymphoma
adopted nomenclature that represents a mixture of clinical (plaque and ulcer) and
histologic (eosinophilic and granuloma) terms.
Considering that the striking clinical phenotype of EGC consists of firm, raised
papules, plaques and nodules and of sharply demarcated ulcers, the main clinical
differential diagnoses include deep bacterial, including mycobacterial, or fungal
infections and neoplasia. Specifically, the main differentials to be taken into account
are squamous cell carcinoma for LU and mast cell tumor, cutaneous lymphocytosis,
and cutaneous infiltration of mammary adenocarcinoma for EP.
In xanthomas, plaques may be whitish-yellow in color, occasionally ulcerated
and occurring on the head and extremities, while they can be hyperkeratotic and
hyperpigmented in papillomas or Bowenoid in situ carcinoma [2, 11]. Erythematous,
eroded, round plaques or nodules clinically indistinguishable from eosinophilic
plaques may be observed in cutaneous lymphocytosis or epitheliotropic cutaneous
lymphoma (Fig. 9) [12, 13].
VetBooks.ir
Plaques, Nodules and Eosinophilic Granuloma Complex Lesions
131
Nodules may be single or multiple. In terms of usefulness for the diagnosis,
relevant clinical features of nodules are location (Table 2), consistency and presence or absence of draining tracts. Soft, fluctuant nodules draining exudate on the
trunk may represent sterile nodular panniculitis or mycobacterial infection
(Fig. 10). A nodule affecting the bridge of the nose and deforming the cat’s profile
(roman nose) may suggest cryptococcosis or nasal lymphoma. Swelling of one or
more footpads may indicate plasma cell pododermatitis (Fig. 11) [14]. Occasionally,
nodules may drain an exudate containing macroscopically visible granules (grains).
The grains are usually white in bacterial botryomycosis, yellow in dermatophytic
mycetomas and of variable colors in eumycotic mycetomas [5]. A nodule in the
Table 2 Common locations
of plaques and nodules in
selected feline skin diseases
Plaques
Distribution
Head, extremities
Abdomen, groin, axillae
Nodules
Abdomen, groin, rump
Abdomen
Head, extremities, tail base
Dorsal nose
Caudal thighs, chin, oral
cavity, paws
Paws
Footpads
Trunk
Pinnae, eyelids, nasal
planum
Abdomen
Interscapular, trunk
Ear canals, pinnae
Fig. 10 Fluctuant nodules
with small ulcers and
draining tract on the flank
and rump due to
mycobacterial infection
(M. smegmatis)
Disease
Xanthomas
Eosinophilic plaque
Rapidly growing
mycobacterial infections
Nocardiosis
Sporotrichosis
Cryptococcosis
Eosinophilic granuloma
Calcinosis cutis
Plasma cell pododermatitis
Sterile nodular panniculitis
Squamous cell carcinoma
Lymphangiosarcoma
Vaccine-site fibrosarcoma
Ceruminous cystomatosis
VetBooks.ir
132
S. Colombo and A. Fondati
Fig. 11 Plasma cell
pododermatitis with
ulceration of the central
metacarpal footpad
interscapular region or dorsolateral thorax should raise suspicion of vaccine-site
fibrosarcoma [8]. Multiple, grey-bluish nodules affecting the face and/or the ear
canals and inner aspect of the pinnae may indicate ceruminous cystomatosis, particularly in Persian cats [3].
Diagnostic Algorithm
This section is illustrated in Figs. 12a, b. Red squares with numbers represent the
steps of the diagnostic process, explained below.
1
Perform cytology
When the lesion is a nodule or a plaque, cytology is the first diagnostic test to
perform during the consultation. Techniques useful to obtain samples for cytological examination from these lesions are fine needle insertion or aspiration and
impression smears, if the nodule is ulcerated or if there is a draining tract.
However, impression smears may be difficult to interpret in “open” lesions due
to potential sample contamination. Cytology allows the clinician to differentiate
between inflammatory and neoplastic infiltrates in most cases, and to select the
most appropriate diagnostic tests to perform thereafter. When a monomorphous
cell population with few or no inflammatory cells is observed, neoplasia should
be suspected. Cytological examination allows to further characterize the cell
population as composed by epithelial, mesenchymal, or round cells, and, in some
cases, it may be diagnostic of a specific neoplasia (e.g., well-differentiated mast
cell tumor). In the majority of cases, however, the lesion must be biopsied or
excised to perform histopathological examination and properly “name” the
tumor.
Plaques, Nodules and Eosinophilic Granuloma Complex Lesions
133
VetBooks.ir
Plaques and nodules 1
Monomorphous cell population
Neoplasia
Epithelial cells
1
Mesenchymal cells
Round cells
2
Non diagnostic
Mast cell tumor
ECL/NECL
Cutaneous lymphocytosis
Melanocytoma
Melanoma
Fibrosarcoma
Hemangiosarcoma
Lymphangiosarcoma
Sarcoid
3
IHC
Molecular techniques
3
Calcium salts
Papillomavirus infections
Histopathology
Squamous cell carcinoma
Basal cell tumor
Follicular tumor
Ceruminous cystomatosis
IHC
Mixed cell population
Inflammation
Plaques and nodules 2
Cytology
3
IHC
Molecular techniques
3
Complete blood count
Biochemistry
Urinalysis
Toluidine blue
IHC
Clonality testing
Calcinosis cutis
Plaques and nodules 2
Monomorphous cell population
Neoplasia
Plaques and nodules 1
Plasma cell pododermatitis
1
Cytology
Mixed cell population
Inflammation
Lymphoplasmacellular
inflammation
Eosinophilic inflammation
Pyogranulomatous/granulomatous inflammation
Suspect:
Leprosy
Cryptococcosis
Sporotrichosis
Leishmaniosis
Pheophyphomycosis
With etiological agent
Suspect:
Botryomycosis
Nocardiosis
Dermatophytic mycetoma
Eumycotic mycetoma
Histopathology
3
Gram stain
Acid fast stains
PCR
Bacterial culture
Bacterial culture and
sensitivity testing
Botryomycosis
Nocardiosis
Go to chapter 9
Pruritus
Without etiological agent
With grains
2
3
Eosinophilic plaque
Eosinophilic granuloma
Sterile nodular panniculitis
Sterile granuloma/pyogranuloma syndrome
Rapidly growing mycobacteria
Leprosy
4
3
Special stains for fungi
Fungal culture
IHC
PCR
Cryptococcosis
Sporotrichosis
Dermatophytic mycetoma
Eumycotic mycetoma
Pheophyphomycosis
Complete blood count
Biochemistry
Urinalysis
Serology
Xanthomatosis
Leishmaniosis
Fig. 12 Diagnostic algorithm of plaques, nodules, and eosinophilic granuloma complex lesions
4
VetBooks.ir
134
2
S. Colombo and A. Fondati
A mixed cell population observed on cytology indicates inflammation.
Inflammatory cells most commonly identified include neutrophils, eosinophils,
macrophages, lymphocytes, plasma cells and mast cells, often accompanied by a
variable amount of red blood cells. The relative percentage of one cell type in
respect to other inflammatory cells is used in cytology to define the different
types of inflammation, such as pyogranulomatous (neutrophils and macrophages
in variable proportions, epithelioid macrophages and giant histiocytic cells),
granulomatous (same as before, with very few or no neutrophils), eosinophilic
and lymphoplasmacellular inflammation. Etiological agents such as bacteria,
fungi, and parasites can also be detected, as well as calcium salts in calcinosis
cutis due to chronic kidney disease. Depending on the type of inflammation and
the microorganism(s) observed, a diagnosis can be made in some cases. For
example, amastigotes of the genus Leishmania in the cytoplasm of macrophages
indicate leishmaniosis, or yeasts of the genus Cryptococcus within pyogranulomatous inflammation are suggestive of cryptococcosis. When the same type of
inflammation is observed with unstained, rod-shaped bacteria within macrophages, mycobacterial diseases should be suspected. Whenever the exudate contains grains, cytology from a squashed grain may be useful: filamentous bacteria
may suggest nocardiosis, while cocci or rods may point toward a diagnosis of
bacterial botryomycosis. If grains appear amorphous and hyphae are detected at
the periphery of a grain, dermatophytic mycetoma or eumycotic mycetoma are
likely diagnoses. All these diagnoses should be confirmed by histopathological
examination and cultures from biopsy samples. Histopathology and culture are
also mandatory in all the cases in which cytology reveals granulomatous or pyogranulomatous inflammation without evidence of etiological agents. A predominantly eosinophilic inflammation together with characteristic clinical findings
points toward a lesion of the EGC, while lymphoplasmacellular inflammation
suggests plasma cell pododermatitis. In case of EGC lesions, the diagnostic
workup is irrespective of the clinical form, the distribution of the lesions, and the
presence or absence of pruritus (Fig. 9b). Histopathological examination may be
performed to confirm the diagnosis.
Cytological examination may also be non-diagnostic, because too few cells
are obtained or the sample is heavily contaminated by blood. In ceruminous cystomatosis, for example, a clear fluid containing variable numbers of macrophages
can be obtained. In these cases, histopathological examination must be carried
out.
Take biopsies for histopathological examination
Histopathological examination is mandatory whenever a neoplastic disease
is suspected. However, a non-neoplastic nodule or plaque also requires, in the
majority of cases, histopathological examination to make or confirm the diagnosis and suggest further diagnostic tests. It must be remembered that the histological appearance of EGC lesions does not always reflect the clinical form and the
eosinophilic infiltrate density is quite variable. LU, for instance, is commonly
reported as a neutrophilic fibrosing dermatitis rather than an eosinophilic dermatitis. A progression of histological lesions has been described from a dermal
VetBooks.ir
Plaques, Nodules and Eosinophilic Granuloma Complex Lesions
3
4
135
eosinophilic infiltrate to fibrosis and neutrophilic ulceration, in a few months, in
the LU of the upper lip. These findings might help to explain why LU is infrequently described as an eosinophil-rich dermatitis. Being clinicians reluctant to
biopsy the cat’s lip, the majority of LU lesions might be present for months at the
time they are histologically examined. In fact, LU are mostly biopsied to rule out
neoplasia rather than to confirm the diagnosis of EGC. When collecting biopsy
samples, some fresh tissue, preferably from the deep portion of the samples,
should be stored in a sterile tube and frozen for possible microbial culture,
molecular studies or both.
Histopathological examination may be diagnostic for neoplasia or, in difficult
cases, additional testing may be needed. Depending on the type of tumor identified or suspected on histopathological examination, special stains (e.g., toluidine
blue or Giemsa for mast cell tumor), immunohistochemistry, or clonality testing
(to differentiate cutaneous lymphocytosis from epitheliotropic cutaneous lymphoma) may be suggested by the pathologist to make the diagnosis.
Special staining, immunohistochemistry and molecular techniques such as
polymerase chain reaction (PCR) may also be useful to identify or characterize
infectious agents which are difficult to see on “standard” histopathology or to
grow on culture. Gram stain is useful to identify bacteria, while acid fast stains
such as Ziehl-Neelsen may be necessary to visualize mycobacteria. Periodicacid of Schiff (PAS) stain is commonly used to identify fungi in tissues.
Immunohistochemistry, PCR and/or other molecular techniques may be applied
to diagnose papillomavirus infections, mycobacterial diseases and some uncommon fungal infections (pheohyphomycosis). If a deep bacterial or fungal infection is suspected, tissue cultures are recommended to identify the causative
microorganisms. Cultures should be preferably performed in specialized
Veterinary Labs, and clinicians should inform of the clinical suspicion. In
selected cases, sensitivity testing can help to choose the correct antimicrobial
treatment. A negative result, together with compatible clinical and histopathological findings, confirms the diagnosis in sterile diseases such as sterile nodular
panniculitis and sterile granuloma/pyogranuloma syndrome.
Perform complete blood count, biochemistry, urinalysis, and serology
Complete blood count, biochemistry, and urinalysis are useful when a metabolic disease such as xanthomas or calcinosis cutis due to renal failure is suspected, based on the results of histopathological examination. If results of
cytology and/or histopathology suggest a diagnosis of leishmaniosis, serology
should also be performed. FIV and FeLV serology should be also carried out,
especially in cats affected by infectious diseases.
References
1. Colombini S, Clay Hodgin E, Foil CS, Hosgood G, Foil LD. Induction of feline flea allergy
dermatitis and the incidence and histopathological characteristics of concurrent lip ulcers. Vet
Dermatol. 2001;12:155–61.
VetBooks.ir
136
S. Colombo and A. Fondati
2. Vogelnest LJ. Skin as a marker of general feline health: cutaneous manifestations of systemic
disease. J Feline Med Surg. 2017;19:948–60.
3. Chaitman J, Van der Voerdt A, Bartick TE. Multiple eyelid cysts resembling apocrine hidrocystomas in three Persian cats and one Himalayan cat. Vet Pathol. 1999;36:474–6.
4. Moriello KA, Coyner K, Paterson S, Mignon B. Diagnosis and treatment of dermatophytosis
in dogs and cats. Clinical consensus guidelines of the world association for veterinary dermatology. Vet Dermatol. 2017;28:266–e68.
5. Backel K, Cain C. Skin as a marker of general feline health: cutaneous manifestations of infectious disease. J Feline Med Surg. 2017;19:1149–65.
6. Gremiao IDF, Menezes RC, Schubach TMP, Figueiredo ABF, Cavalcanti MCH, Pereira
SA. Feline sporotrichosis: epidemiological and clinical aspects. Med Mycol. 2015;53:15–21.
7. Pennisi MG, Cardoso L, Baneth G, Bourdeau P, Koutinas A, Mirò G, Oliva G, Solano-­
Gallego L. LeishVet update and recommendations on feline leishmaniosis. Parasit Vectors.
2015;8:302–20.
8. Hartmann K, Day MJ, Thiry E, Lloret A, Frymus T, Addie D, Boucraut-Baralon C, Egberink
H, Gruffydd-Jones T, Horzinek MC, Hosie MJ, Lutz H, Marsilio F, Pennisi MG, Radford AD,
Truyen U, Möstl K. Feline injection-site sarcoma: ABCD guidelines on prevention and management. J Feline Med Surg. 2015;17:606–13.
9. Buckley L, Nuttall T. Feline eosinophilic granuloma complex(ities) some clinical clarification.
J Feline Med Surg. 2012;14:471–81.
10. Wildermuth BE, Griffin CE, Rosenkrantz WS. Response of feline eosinophilic plaques and lip
ulcers to amoxicillin trihydrate–clavulanate potassium therapy: a randomized, double-blind
placebo-controlled prospective study. Vet Dermatol. 2011;23:110–e25.
11. Munday JS. Papillomaviruses in felids. Vet J. 2014;199:340–7.
12. Gilbert S, Affolter VK, Gross TL, Moore PF, Ihrke PJ. Clinical, morphological and immunohistochemical characterization of cutaneous lymphocytosis in 23 cats. Vet Dermatol.
2004;15:3–12.
13. Fontaine J, Heimann M, Day MJ. Cutaneous epitheliotropic T-cell lymphoma in the cat: a
review of the literature and five new cases. Vet Dermatol. 2011;22:454–61.
14. Dias Pereira P, Faustino AMR. Feline plasma cell pododermatitis: a study of 8 cases. Vet
Dermatol. 2003;14:333–7.
Further Readings
“Plaque, nodule”. Merriam-Webster Medical Dictionary. http://merriam-webster.com Accessed 31
Jan 2018.
Albanese F. Canine and feline skin cytology. Cham: Springer International Publishing; 2017.
Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, Wolff K. Fitzpatrick’s dermatology in
general medicine. 8th ed. New York: The McGraw-Hill Companies; 2012.
Gross TL, Ihrke PJ, Walder EJ, Affolter VK. Skin diseases of the dog and cat. Clinical and histopathologic diagnosis. 2nd ed. Oxford: Blackwell Publishing; 2005.
Miller WH, Griffin CE, Muller CKL. Kirk’s small animal dermatology. 7th ed. St. Louis: Elsevier;
2013.
Noli C, Toma S. Dermatologia del cane e del gatto. 2nd ed. Vermezzo: Poletto Editore; 2011.
VetBooks.ir
Excoriations, Erosions and Ulcers
Silvia Colombo
Abstract
Excoriations, erosions and ulcers are relatively common lesions in the cat, and in
general, they are quite non-specific. Excoriations are, by definition, self-­induced
lesions due to scratching, while erosions and ulcers develop spontaneously. Full
thickness skin wounds are very suggestive of cutaneous asthenia or acquired skin
fragility syndrome, depending on the cat’s age. Erosions and ulcers are often
secondarily infected and may appear more severe because of pruritus due to
infection. A peculiar feline clinical presentation, common in allergic diseases, is
“head and neck pruritus,” with excoriations and ulcers being self-induced. This
presentation is usually investigated following the diagnostic approach to pruritus. The most relevant clinical feature of erosions and ulcers is their location,
which may be helpful for the diagnosis. In general, histopathology is the most
important diagnostic test to make a specific diagnosis in erosive/ulcerative feline
skin diseases.
Definitions
An excoriation is a superficial abrasion of the epidermis that results from scratching, or, less commonly, from licking or biting. It is a self-induced lesion and may
show a linear pattern, directly reflecting its pathogenesis.
An erosion is a superficial, moist, circumscribed lesion that results from loss of
a part or all of the epidermis and does not involve the dermis. An erosion does not
bleed and heals without scarring.
An ulcer is a circumscribed skin defect in which the epidermis and at least the
superficial dermis have been lost, and it is deeper than the erosion. The ulcer involves
S. Colombo (*)
Servizi Dermatologici Veterinari, Legnano, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_7
137
VetBooks.ir
138
S. Colombo
also the adnexa and may heal with scarring. Further features used to describe an
ulcer relate to its margins, surface and presence of exudate eventually covering its
bottom. The margins, for example, may be thickened, regular or irregular, and the
bottom may be clean, hemorrhagic or necrotic. There may be a crust or purulent
exudate covering the ulcerated area.
Erosion and ulcer are difficult to be differentiated clinically, because the depth of
a skin defect can only be defined with certainty by histopathological examination.
For this reason, when describing a typical lesion or a disease, the words erosive and
ulcerative are always used together.
Pathogenesis
The pathogenetic mechanisms underlying the formation of erosions and ulcers
vary from external trauma, to congenital defects causing reduced skin resistance,
to direct infectious or autoimmune damage to the skin. Erosions and ulcers are, in
the vast majority of cases, complicated by self-induced trauma due to pruritus and/
or by secondary infections. The clinical appearance of any erosive and ulcerative
disease may therefore evolve, and lesions may become deeper and more severe.
A peculiar feline clinical presentation, common in allergic diseases, is ulceration
due to “head and neck pruritus” (Fig. 1) [1]. Cats scratch using their hind paws and
claws and may cause severe and extensive ulcers to these locations.
Despite its name, the feline-specific lesion known as “indolent ulcer” or “lip
ulcer” (Fig. 2) is an ulcerated plaque and is described in Chapter, Plaques, Nodules
and Eosinophilic Granuloma Complex Lesions [2].
Fig. 1 Head and neck
pruritus in an allergic cat
VetBooks.ir
Excoriations, Erosions and Ulcers
139
Fig. 2 Severe, bilateral
indolent ulcer with necrotic
material in the center
In diseases such as cutaneous asthenia or acquired skin fragility syndrome, full
thickness lacerations and skin detachment occur following minor trauma, and the
lesions should be better described as wounds. Table 1 lists selected causes of excoriations, erosions and ulcers in cats.
Diagnostic Approach
Signalment and History
Erosive/ulcerative skin diseases such as cutaneous asthenia and dystrophic or junctional epidermolysis bullosa are congenital and present at birth or shortly thereafter
[3, 4]. In other cases, the disease has a delayed onset but is clinically apparent
in young adult cats of specific breeds (idiopathic facial dermatitis of Persian and
Himalayan cats, ulcerative nasal dermatitis of Bengal cats) [5, 6]. Full thickness
wounds following minor trauma may occur in senior to geriatric cats in acquired
skin fragility syndrome, which may be caused by hyperadrenocorticism (Fig. 3) or
other diseases [7, 8]. Traumatic excoriations or wounds may be seen more often in
tomcats, while neoplastic diseases are more common in older cats.
History is very relevant for the diagnosis of feline erosive/ulcerative diseases.
Previous or concurrent respiratory clinical signs may suggest herpesvirus dermatitis, while an outdoor lifestyle may predispose the cat to trauma, deep bacterial,
mycobacterial or fungal infections, or, in white cats, to squamous cell carcinoma
(Fig. 4) [9, 10]. Previous or concurrent drug administration should prompt the clinician to include adverse drug reactions and toxic epidermal necrolysis among the
differential diagnoses, particularly if the lesions have a sudden onset [11]. Finally,
the presence of pruritus may be a relevant information as it may be typical of some
140
S. Colombo
Table 1 Selected causes of excoriations, erosions and ulcers
VetBooks.ir
Excoriations
Erosions/ulcers
Self-trauma
Herpesvirus dermatitis
Leprosy
Rapidly growing mycobacterial infections
Subcutaneous fungal infections
Systemic fungal infections
Myiasis
Leishmaniosis
Head and neck pruritusa (Table 3)
Adverse drug reactions
Pemphigus foliaceus
Pemphigus vulgaris
Vesicular diseases of the dermo-epidermal junction
Erythema multiforme
Toxic epidermal necrolysis
Vasculitis
Hyperadrenocorticism/acquired skin fragility syndrome
Idiopathic facial dermatitis of Persian and Himalayan cats
Ulcerative nasal dermatitis of Bengal cats
Junctional/dystrophic epidermolysis bullosa
Cutaneous asthenia
Trauma
Indolent ulcerb
Idiopathic/behavioral ulcerative dermatitis
Plasma cell pododermatitis
Squamous cell carcinoma
Chapter, Pruritus
Chapter, Plaques, Nodules and Eosinophilic Granuloma Complex Lesions
a
b
Fig. 3 Full thickness skin
wound in a geriatric cat
with hyperadrenocorticism
VetBooks.ir
Excoriations, Erosions and Ulcers
141
Fig. 4 Squamous cell
carcinoma involving the
lower eyelid and nose of a
white cat
diseases such as idiopathic/behavioral ulcerative dermatitis and of the clinical pattern described as “head and neck pruritus” [1, 12]. However, one must remember
that pruritus may also be due to secondary infections of the erosion/ulcer.
Clinical Presentation
Erosions and ulcers are relatively common lesions in the cat and are quite non-­
specific. Accompanying primary or secondary lesions are uncommon, with the
exception of crusts covering the erosions/ulcers. Nodules and plaques may have an
eroded or ulcerated surface, as it happens in indolent ulcer and eosinophilic plaque.
On the other hand, full thickness wounds are very specific once a traumatic etiology has been ruled out. Skin tears with minimal or no bleeding, occurring following minor traction, suggest cutaneous asthenia or acquired skin fragility syndrome,
depending on the patient’s age [3, 7, 8, 13]. The concurrent presence of skin hyperextensibility is a feature of cutaneous asthenia, while thin, irregular scars represent
resolved lesions and may be observed in both conditions.
The most useful clinical features of erosions/ulcers are the lesions’ location
(Table 2) and the presence or absence of pruritus. The face is the most common site
for erosions/ulcers due to herpesvirus (Fig. 5) or calicivirus infections, occasionally with oral cavity involvement [9, 10]. Idiopathic facial dermatitis of Persian
and Himalayan cats is initially characterized by an accumulation of adherent black
material around the eyes, nose and mouth, and inflamed, eroded/ulcerated skin
lesions underneath the exudate develop with time [5]. These lesions may be severely
pruritic and secondary infections are common. Multiple erosions/ulcers, covered by
crusts, on the tip of the pinnae, eyelids and/or nasal planum of a white cat should
prompt the clinician to investigate squamous cell carcinoma. A hyperkeratotic,
scaly, occasionally ulcerated nasal planum in Bengal cats has been reported and is
thought to be a congenital disease [6]. Application of spot-on products to prevent
142
S. Colombo
VetBooks.ir
Table 2 Common locations of erosions/ulcers in selected feline skin diseases
Distribution
Oral cavity
Abdomen, groin
Upper lip
Dorsal neck
Footpads
Trunk
Nasal planum
Muzzle
Pinnae
Eyelids
Fig. 5 Large erosion/ulcer
on the muzzle of a cat with
herpesvirus infection
Disease
Herpesvirus dermatitis
Pemphigus vulgaris
Vesicular diseases of the dermo-epidermal junction
Rapidly growing mycobacterial infections
Eosinophilic plaques
Indolent ulcer
Adverse drug reaction (spot-on, injection)
Idiopathic/behavioral ulcerative dermatitis
“Head and neck pruritus”
Plasma cell pododermatitis
Hyperadrenocorticism
Acquired skin fragility syndrome
Ulcerative nasal dermatitis of Bengal cats
Squamous cell carcinoma
Pemphigus foliaceus
Idiopathic facial dermatitis of Persian and Himalayan cats
Pemphigus foliaceus
Herpesvirus dermatitis
“Head and neck pruritus”
Squamous cell carcinoma
Pemphigus foliaceus
Squamous cell carcinoma
Pemphigus vulgaris
Vesicular diseases of the dermo-epidermal junction
VetBooks.ir
Excoriations, Erosions and Ulcers
143
ectoparasites may cause erosions/ulcers on the dorsal neck. Ulcerative lesions and
draining tracts discharging exudate on the abdomen may be observed in rapidly
growing mycobacterial infections [10]. Severely swollen, ulcerated metacarpal and/
or metatarsal footpads are suggestive of plasma cell pododermatitis [14]. In erythema multiforme, maculopapular lesions evolving to erosions/ulcers and crusts
usually present with a generalized distribution [11].
ead and Neck Pruritus
H
Pruritus and self-induced erosions/ulcers involving the head, pinnae and neck are
commonly observed and represent a peculiar clinical presentation in the feline species [1]. Variably sized erosions and ulcers are caused by the cat’s scratching with
the hind paws, and the owner is usually well aware of the cat’s pruritus. These
lesions may be very severe, secondary infections are often present, and their depth
may reach the subcutis (Fig. 6). Other feline presentations typically observed in
pruritic skin diseases, such as miliary dermatitis and self-induced alopecia, may be
concurrently observed. Differential diagnoses of head and neck pruritus are listed
in Table 3. Head and neck pruritus should be investigated following the diagnostic
approach to pruritus (Chapter, Pruritus).
Idiopathic/behavioral ulcerative dermatitis (Fig. 7), which presents as a very
severe and extremely pruritic, usually single crusted ulceration affecting the dorsal neck, deserves a specific comment, since its etiopathogenesis is controversial.
Suggested causes of idiopathic ulcerative dermatitis involve allergic diseases,
Fig. 6 Very severe
erosions/ulcers in a food
allergic cat
VetBooks.ir
144
S. Colombo
Table 3 Differential
diagnoses of head and neck
pruritus
Disease
Herpesvirus dermatitis
Dermatophytosis
Notoedric mange
Otodectic mange
Demodicosis (Demodex gatoi)
Trombiculiasis
Lynxacarus infestation
Flea-bite hypersensitivity
Adverse reaction to food
Feline atopic syndrome
Mosquito-bite hypersensitivity
Adverse drug reaction
Pemphigus foliaceus
Idiopathic facial dermatitis of Persian and Himalayan
cats
Idiopathic/behavioral ulcerative dermatitis
Fig. 7 Idiopathic/
behavioral ulcerative
dermatitis on the dorsal
neck
secondary infections, neurological diseases and a behavioral disorder, although
most cases, as the name suggests, are idiopathic [12].
Diagnostic Algorithm
This section is illustrated in Fig. 8. Red squares with numbers represent the steps of
the diagnostic process, as explained below.
1
Consider history and clinical examination.
When examining a cat with erosions/ulcers, the first step is to separate these
lesions from full thickness wounds or skin lacerations following minor trauma,
Excoriations, Erosions and Ulcers
145
VetBooks.ir
Erosions and ulcers
History and clinical examination 1
Full thickness wound
Head and neck pruritus
Indolent ulcer
Monomorphous cell population
Neoplasia
Cytology
Mixed cell population
Inflammation
2
3 Histopathology
Histopathology
3
Trauma
Myasis
Non diagnostic
Neutrophilic inflammation, erythrocytes
Acquired skin fragility syndrome Cutaneous asthenia
Neutrophilic inflammation
Acantholytic cells
Pyogranulomatous/granulomatous
inflammation
Lymphoplasmacellular
inflammation
Squamous cell carcinoma
Epitheliotropic cutaneous lymphoma
3 Histopathology
3 Histopathology
Plasma cell pododermatitis
Pemphigus foliaceus
Vesicular diseases of the dermo-epidermal junction
Adverse drug reaction
Erythema multiforme
Toxic epidermal necrolysis
Idiopathic facial dermatitis of Persian and Himalayan cats
Ulcerative nasal dermatitis of Bengal cats
Junctional/dystrophic epidermolysis bullosa
4
Complete blood count
Biochemistry
Urinalysis
Endocrinology testing
X-ray
Ultrasound
TC/MRI
(if appropriate)
Leprosy
Rapidly growing mycobacterial infections
Subcutaneous fungal infections
Systemic fungal infections
Leishmaniosis
4
Complete blood count
Biochemistry
Urinalysis
Serology
PCR
Go to chapter 9
Pruritus
Leishmaniosis
Special stains
Fungal culture
Bacterial culture
IHC
PCR
5
Leprosy
Rapidly growing mycobacterial infections
Subcutaneous fungal infections
Systemic fungal infections
Fig. 8 Diagnostic algorithm of erosions and ulcers
2
such as manual traction. Extreme fragility of the skin only occurs in two conditions, in cats. The first one is cutaneous asthenia, clinically apparent in kittens
or young cats, while the second one occurs in old felines and gathers different
diseases under the name “acquired skin fragility syndrome” [3, 7, 8, 13]. History
would also help us, in most cases, to decide if the wound occurred following a
major trauma such as a car accident. The presence of insect larvae in the wound
indicates myiasis. If the cat is pruritic on the head and neck, or the main lesion
is the “indolent” lip ulcer or an eroded plaque (Chapter, Plaques, Nodules and
Eosinophilic Granuloma Complex Lesions), particularly when associated with
self-induced alopecia, one should follow the diagnostic approach to pruritus,
described in Chapter, Pruritus.
Perform cytology.
Cytologic examination of erosions/ulcers is often disappointing, because in
most cases one can only observe non-specific findings, such as red blood cells
and neutrophils. When neutrophils are present admixed with acantholytic cells,
the main clinical suspicion is pemphigus foliaceus (Fig. 9). If a mixed cell population comprising large numbers of plasma cells and lymphocytes are observed and
the cytology sample comes from a footpad, the diagnosis is plasma cell pododermatitis [14]. Eosinophils may be observed on samples taken from the tiny erosions underneath the crusts in miliary dermatitis, or from the eroded surface of an
eosinophilic plaque. Pyogranulomatous inflammation is also a non-­
specific
VetBooks.ir
146
S. Colombo
Fig. 9 Pemphigus
foliaceus in a domestic
short-haired cat
3
4
5
c­ ytological picture; however, it is more commonly observed in infectious diseases
such as mycobacterial or fungal infections and leishmaniosis. O
­ ccasionally, a
monomorphous cell population is seen on cytology, and this finding may suggest
a neoplastic disease.
Perform histopathology.
Histopathology is of paramount importance in erosive/ulcerative feline skin
diseases. First of all, it can confirm the diagnosis of neoplasia and acquired skin
fragility syndrome. In cutaneous asthenia, histopathological comparison with a
skin sample obtained from a cat of the same age and from the same site may be
required, as well as special stains and electron microscopy. The majority of
autoimmune, immune-mediated and idiopathic erosive/ulcerative skin diseases
can be diagnosed by histopathology. In case of infectious diseases, a standard
histopathological examination (hematoxylin-eosin, H&E) is in most cases only
indicative, due to the difficulty of identifying the etiological agent without further diagnostic procedures, such as special stains or immunohistochemistry.
Perform blood testing, serology, urinalysis and diagnostic imaging.
In an old cat presenting with full thickness wounds and a histopathological
diagnosis of acquired skin fragility syndrome, it is necessary to identify the
causative disease in order to attempt a treatment. Skin fragility syndrome is
often caused by hyperadrenocorticism; however, severe cachexia, diabetes mellitus, hepatic lipidosis or inflammatory and neoplastic diseases affecting the
liver, nephrosis and some infectious diseases have all been reported [8, 13].
When history, clinical examination and histopathology suggest leishmaniosis,
the diagnostic process should be completed by complete blood count, biochemistry, urinalysis, serology and/or PCR [10].
As said before, standard histopathology (H&E staining) may in some cases be
suggestive of an infectious disease, without confirming a specific diagnosis.
In these cases, further testing is mandatory and special stains (PAS for fungi,
Ziehl-­Neelsen for acid-fast bacteria, immunohistochemistry for leishmania
and viruses), cultures and PCR should be requested depending on the case.
Excoriations, Erosions and Ulcers
147
VetBooks.ir
References
1. Hobi S, Linek M, Marignac G, Olivry T, et al. Clinical characteristics and causes of pruritus
in cats: a multicentre study on feline hypersensitivity-associated dermatoses. Vet Dermatol.
2011;22:406–13.
2. Buckley L, Nuttall T. Feline Eosinophilic Granuloma Complex(ITIES): some clinical clarification. J Fel Med Surg. 2012;14:471–81.
3. Hansen N, Foster SF, Burrows AK, Mackie J, Malik R. Cutaneous asthenia (Ehlers–Danlos-­
like syndrome) of Burmese cats. J Feline Med Surg. 2015;17:954–63.
4. Medeiros GX, Riet-Correa F. Epidermolysis bullosa in animals: a review. Vet Dermatol.
2015;26:3–e2.
5. Bond R, Curtis CF, Ferguson EA, Mason IS, Rest J. An idiopathic facial dermatitis of Persian
cats. Vet Dermatol. 2000;11:35–41.
6. Bergvall K. A novel ulcerative nasal dermatitis of Bengal cats. Vet Dermatol. 2004;15:28.
7. Boland LA, Barrs VR. Peculiarities of feline hyperadrenocorticism: update on diagnosis and
treatment. J Feline Med Surg. 2017;19:933–47.
8. Furiani N, Porcellato I, Brachelente C. Reversible and cachexia-associated feline skin fragility
syndrome in three cats. Vet Dermatol. 2017;28:508–e121.
9. Hargis AM, Ginn PE. Feline herpesvirus 1-associated facial and nasal dermatitis and stomatitis
in domestic cats. Vet Clin North Am Small Anim Pract. 1999;29(6):1281–90.
10. Backel K, Cain C. Skin as a marker of general feline health: cutaneous manifestations of infectious disease. J Feline Med Surg. 2017;19:1149–65.
11. Yager JA. Erythema multiforme, Stevens–Johnson syndrome and toxic epidermal necrolysis:
a comparative review. Vet Dermatol. 2014;25:406–e64.
12. Titeux E, Gilbert C, Briand A, Cochet-Faivre N. From feline idiopathic ulcerative dermatitis
to feline behavioural ulcerative dermatitis: grooming repetitive behaviors indicators of poor
welfare in cats. Front Vet Sci. 2018; https://doi.org/10.3389/fvets.2018.00081.
13. Vogelnest LJ. Skin as a marker of general feline health: cutaneous manifestations of systemic
disease. J Feline Med Surg. 2017;19:948–60.
14. Dias Pereira P, Faustino AMR. Feline plasma cell pododermatitis: a study of 8 cases. Vet
Dermatol. 2003;14:333–7.
General References
For definitions: Merriam-Webster Medical Dictionary. http://merriam-webster.com Accessed 10
May 2018.
Albanese F. Canine and feline skin cytology. Cham: Springer International Publishing; 2017.
Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, Wolff K. Fitzpatrick’s dermatology in
general medicine. 8th ed. New York: The McGraw-Hill Companies; 2012.
Miller WH, Griffin CE, Campbell KL. Muller & Kirk’s Small Animal Dermatology. 7th ed. St.
Louis: Elsevier; 2013.
Noli C, Toma S. Dermatologia del cane e del gatto. 2nd ed. Vermezzo: Poletto Editore; 2011.
VetBooks.ir
Scaling
Silvia Colombo
Abstract
Exfoliative diseases in cats are clinically characterized by dry or greasy scaling
and, less commonly, by follicular casts. In normal skin, there is a continuous
turnover of cells, with new keratinocytes being produced in the basal layer and
migrating upward to become non-nucleated corneocytes in the stratum corneum.
Corneocytes are shed in the environment and are not visible to the naked eye.
When this process is abnormal, scales become macroscopically visible. The
most common cause of scaling in cats is poor grooming, usually associated with
older age, obesity or concurrent systemic diseases. Greasy scaling is often associated with Malassezia overgrowth, while follicular casts are rare in the feline
species. The diagnostic approach involves ruling out ectoparasitic diseases and
dermatophytosis, evaluating the presence or absence of Malassezia spp. by cytology, and assessing the cat’s general health status, especially in older patients.
Histopathology is usually required to make the diagnosis of the majority of exfoliative dermatoses.
Definitions
A scale is a small, thin, dry piece of cornified layer detaching from the skin, and
scaling is the process of shedding scales. In English, scale and squame as well as
scaling, desquamation and exfoliation are synonyms and are used indifferently. The
informal term used to describe scaling is dandruff. In normal conditions, exfoliation occurs continuously, without the formation of visible scaling. Desquamation
becomes visible when it occurs in increased amount, because the epidermal differentiation is abnormal.
S. Colombo (*)
Servizi Dermatologici Veterinari, Legnano, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_8
149
VetBooks.ir
150
S. Colombo
Scales may be further characterized as dry or greasy, and their color may be
white, silver, yellow, brown or grey, depending on the causative disease. Dry
scaling is common in cats, while greasy scaling is observed only in a few skin
diseases. Scales are also often described as pityriasiform, which means small,
thin, whitish and similar to oat bran, or psoriasiform, a term used to describe
larger, thicker and often silvery scales. Scales arranged in a circle are described as
an epidermal collarette and are rarely observed in cats. The epidermal collarette
is the final evolutive stage of a papule or a pustule (Chapter, Papules, Pustules,
Furuncles and Crusts).
A follicular cast is an accumulation of keratin and follicular content which
adheres to the hair shaft, protruding from the follicular ostium. This material often
glues together a tuft of hair or may accumulate around a single hair shaft. Follicular
casts are very uncommon in cats but may represent a useful clinical hint toward the
diagnosis.
Pathogenesis
In the normal skin, there is a continuous turnover of cells, with new keratinocytes
being produced in the basal layer, maturing in the spinous layer, and dying to
become corneocytes in the horny layer. Corneocytes are shed in the environment
and are not visible to the naked eye. In abnormal situations, scaling becomes obvious because the corneocytes detach in larger clusters. This may be due to increased
production or reduced shedding of the horny layer or to abnormalities of the superficial lipid film, that covers and protects the skin surface. An increased production of
the horny layer may occur in congenital diseases such as ichthyosis or primary seborrhea; however, these conditions are extremely rare in cats [1]. More commonly,
the increased thickness of the horny layer is a response to an external insult, such
as sunrays damage in solar dermatitis (Fig. 1), which may evolve into actinic keratosis and squamous cell carcinoma, or to ectoparasites feeding on the skin surface
in cheyletiellosis. Another pathogenetic mechanism underlying scaling dermatoses
is the infiltration of inflammatory or neoplastic cells in the skin, as it happens in
erythema multiforme (Fig. 2), exfoliative dermatitis with/without thymoma or epitheliotropic cutaneous lymphoma [2–4].
In cats, reduced shedding of horny layer is usually due to poor grooming, more
commonly in older or obese cats or in cats affected by systemic diseases such as
diabetes mellitus or hyperthyroidism. The lipid film that protects the skin is, at least
in part, produced by the sebaceous glands. Diseases affecting and destroying these
glands, such as sebaceous adenitis or leishmaniosis, may present with scaling [3, 5].
Although very uncommonly, scaling in cats may also be greasy. This may occur
due to excessive production of glandular secretions in primary seborrhea, a very
rare disease in cats, and in the more common tail gland hyperplasia, also known as
VetBooks.ir
Scaling
151
Fig. 1 Scaling, erythema,
and mild crusting on the
pinna of a white cat with
solar dermatitis
Fig. 2 Scaling on the
footpads of a cat affected
by erythema multiforme
stud tail (Fig. 3). Malassezia overgrowth may be related to sebaceous gland hyperplasia and abnormalities of the superficial lipid film, and consequently presents with
greasy scaling [6, 7].
Follicular casting is uncommon in the feline species. It may be the clinical expression of follicular damage, as in demodicosis and dermatophytosis, or of destruction
of the sebaceous glands in sebaceous adenitis [3]. A rare, recently reported congenital disease called sebaceous gland dysplasia occurs in kittens and is clinically
characterized by generalized hypotrichosis, scaling and follicular casts [8]. Selected
feline diseases presenting with scaling are listed in Table 1.
VetBooks.ir
152
S. Colombo
Fig. 3 Greasy seborrhea
on the dorsal tail of a
Persian cat with tail gland
hyperplasia
Diagnostic Approach
Signalment and History
Exfoliative diseases such as dermatophytosis (Fig. 4) or cheyletiellosis are commonly observed in kittens or in environmental conditions of crowding, such as
breeding colonies or pet shops. Congenital diseases presenting with dry or greasy
scaling and/or follicular casting are observed in kittens, although primary seborrhea and sebaceous gland dysplasia are extremely rare diseases [2, 9]. The most
common cause of dry scaling in senior and geriatric cats is poor grooming, which
may be due to obesity (Fig. 5) or concurrent systemic diseases such as chronic
renal insufficiency, hyperthyroidism or diabetes mellitus. Less commonly, aged
VetBooks.ir
Scaling
Table 1 Selected diseases
presenting with dry or greasy
scaling and follicular casts in
cats
153
Poor grooming due to obesity or systemic disease
Cheyletiellosis
Demodicosis
Dermatophytosis
Malassezia overgrowth
Leishmaniosis
Adverse drug reaction
Erythema multiforme
Sebaceous gland dysplasia
Primary seborrhea
Sebaceous adenitis
Plasma cell pododermatitis
Solar dermatitis
Tail gland hyperplasia
Exfoliative dermatitis (thymoma-­associated or not)
Epitheliotropic cutaneous lymphoma
Fig. 4 Scaling and
erythema on the margin of
the pinna of a kitten
affected by
dermatophytosis
cats may be affected by neoplastic diseases or paraneoplastic syndromes [4, 9].
Dermatophytosis should always be considered in a Persian cat presenting with
scaling and alopecia, regardless of age and lifestyle (Fig. 6). White cats and cats
with white ears and/or muzzle are predisposed to solar dermatitis, if they are
allowed outdoors and like laying in the sun.
History in adult to older cats should always include concurrently or previously
administered drugs which may cause adverse drug reactions. Finally, in a cat
presenting with scaling, FIV and FeLV statuses should be evaluated. A FeLV-­
associated giant cell dermatosis has been reported to cause generalized, severe
exfoliation, and both viral infections may predispose the cat to other infectious
diseases [10].
VetBooks.ir
154
S. Colombo
Fig. 5 Mild generalized
scaling in a geriatric and
obese cat
Fig. 6 Focal alopecia and
scaling in a Persian cat
with dermatophytosis
Clinical Presentation
Dry scaling of variable severity is common in cats, and further clinical features
should be considered to help listing the differential diagnoses. Generalized or
dorsally distributed pityriasiform scaling in an older cat may be simply due to
poor grooming, while in a recently acquired kitten, it may suggest cheyletiellosis,
especially if pruritus is also reported. Exfoliative erythroderma, a clinical presentation characterized by scaling, erythema, and often alopecia, has been reported in
senior or geriatric cats affected by epitheliotropic cutaneous lymphoma, although
the disease is rare in the cat [4]. Exfoliative dermatoses in cats are often generalized, with a few exceptions. When scaling is associated to focal or multifocal
alopecia, dermatophytosis is a possible diagnosis. Mild scaling and erythema on
the pinnae of a white cat should prompt the clinician to include solar dermatitis
in the differentials.
VetBooks.ir
Scaling
155
Fig. 7 Severe
psoriasiform scaling in
thymoma-associated
exfoliative dermatitis
Psoriasiform scaling is uncommon in cats. In middle-age to older cats, non-­
pruritic, severe, generalized, psoriasiform scaling with a history of starting on the
head and neck and associated alopecia and erythema may be suggestive of thymoma
or non-thymoma-associated exfoliative dermatitis (Fig. 7) [9, 11]. In thymoma-­
associated cases, coughing, dyspnea, depression, anorexia and weight loss are
usually observed after the skin lesions. Psoriasiform scaling, follicular casting
and alopecia with a generalized distribution, associated with deposition of darkcolored debris on the eyelids, may be consistent with sebaceous adenitis (Fig. 8),
an extremely rare disease in the cat [3]. Scaling limited to the footpads may be a
clinical feature of plasma cell pododermatitis (Fig. 9) [12].
Localized or generalized greasy scaling, erythema, pruritus and rancid smell
may suggest Malassezia overgrowth (Fig. 10). This disease may be observed both
in young, allergic cats and in older felines with severe systemic diseases, neoplasia
or paraneoplastic syndromes [6, 7]. Greasy scaling on the dorsal aspect of the tail is
the clinical presentation of tail gland hyperplasia, also known as stud tail.
Diagnostic Algorithm
This section is illustrated in Fig. 11. Red squares with numbers represent the steps
of the diagnostic process, as explained below.
1
Perform skin scrapings and microscopic examination of scales and skin debris.
The diagnostic approach to scaling in cats begins with simple tests to diagnose or rule out ectoparasites. Multiple skin scrapings should be performed to
diagnose or rule out demodicosis, and fungal spores can also be seen surrounding and invading fragments of hair shafts in dermatophytosis. Cheyletiellosis
may also be diagnosed with skin scrapings, although the most commonly used
test is microscopic examination of acetate tape strips, after collecting scales
VetBooks.ir
156
Fig. 8 Severe exfoliation
and alopecia on the ventral
trunk of a cat affected by
sebaceous adenitis
Fig. 9 Scaling on the
footpads in a mild case of
plasma cell pododermatitis
S. Colombo
VetBooks.ir
Scaling
157
Fig. 10 Greasy, brown
scaling in the interdigital
spaces of an allergic Devon
Rex cat with Malassezia
overgrowth
Scaling
1
Skin scraping
Microscopic examination
of scales and skin debris
Demodicosis
Cheyletiellosis
Wood’s lamp examination
Fungal culture
Dermatophytosis
Plasma cell pododermatitis
Tail gland hyperplasia
Obesity
3
Cytology
2
Malassezia overgrowth
Signalment, History, Physical examination
Poor grooming
6
4
Histopathology
5
Primary seborrhea
Sebaceous gland dysplasia
Solar dermatitis
Sebaceous adenitis
Erythema multiforme
Adverse drug reactions
Epitheliotropic cutaneous lymphoma
Complete blood count
Biochemistry
Urinalysis
FIV FeLV serology
Leishmania serology
Endocrinology testing
X-ray
Ultrasound
TC/MRI
(if appropriate)
Go to chapter 9
Pruritus
Diabetes mellitus
Chronic kidney disease
Hyperthyroidism
Thymoma-associated exfoliative dermatitis
Non thymoma-associated exfoliative dermatitis
FeLV-associated giant cell dermatosis
Leishmaniosis
Fig. 11 Algorithm of the approach to feline scaling
2
directly from the cat’s coat or from material collected from the examination
table after vigorous stroking of the coat.
Perform Wood’s lamp examination and fungal culture.
These two diagnostic tests, taken together, are diagnostic for dermatophytosis or, if negative results are obtained, are helpful to rule it out. Since dermatophytosis is common in cats, a fungal culture is appropriate in all cases presenting
with scaling.
158
VetBooks.ir
3
4
5
6
S. Colombo
Perform cytology.
Cytology is particularly useful when greasy scaling is a presenting sign.
Samples may be taken by impression smear, using a cotton swab or a piece of
acetate tape to look for Malassezia yeasts. Since these yeasts may be identified
in both young, allergic cats and older felines with systemic diseases, neoplasia
or paraneoplastic syndromes, further investigations should always be carried
out based on signalment, history and clinical examination. When a scaly footpad has to be sampled because plasma cell pododermatitis is suspected, fine
needle capillary suction and aspiration are the preferred techniques.
Consider the patient’s signalment, history and physical examination.
In a young to adult cat presenting with greasy scaling on the dorsal tail, after
ruling out ectoparasitic and fungal diseases, the diagnosis of tail gland hyperplasia is straightforward. When the patient is a senior or geriatric cat and presents with generalized dry scaling, poor grooming is a major differential. An
aged cat may groom with difficulty because it is obese or because it suffers from
a metabolic disease. Depending on other clinical signs, when identified on general physical examination, a variety of diagnostic tests may be appropriate.
Perform blood testing, urinalysis and diagnostic imaging.
In an older cat, basic information should always be obtained by taking a
blood sample and a urine sample for complete blood count, biochemistry, urinalysis and serum total thyroxine (T4) concentrations. This will be useful also
if sedation or general anesthesia is planned for biopsies. FIV and FeLV serology
must be carried out if a cutaneous disease linked to one of these viruses is suspected, although this may become obvious only after histopathological examination. The same applies to serology for leishmaniosis, a rare disease in cats.
Thoracic radiography and/or CT/MRI may be diagnostic for thymoma, which is
often associated with exfoliative dermatitis.
Take biopsies for histopathological examination.
Histopathological examination usually confirms the diagnosis, whether the
clinician is facing a congenital disease or an acquired one. Figure 11 summarizes the most important exfoliative disorders which require biopsies for the
diagnosis.
References
1. Paradis M, Scott DW. Hereditary primary seborrhea oleosa in Persian cats. Feline Pract.
1990;18:17–20.
2. Yager JA. Erythema multiforme, Stevens-Johnson syndrome and toxic epidermal necrolysis: a
comparative review. Vet Dermatol. 2014;25:406–e64.
3. Noli C, Toma S. Case report three cases of immune-mediated adnexal skin disease treated with
cyclosporin. Vet Dermatol. 2006;17(1):85–92.
4. Fontaine J, Heimann M, Day MJ. Cutaneous epitheliotropic T-cell lymphoma in the cat : a
review of the literature and five new cases. Vet Dermatol. 2011;22(5):454–61.
VetBooks.ir
Scaling
159
5. Pennisi MG, Cardoso L, Baneth G, Bourdeau P, Koutinas A, Miró G, et al. LeishVet update
and recommendations on feline leishmaniosis. Parasit Vectors. 2015;8:1–18.
6. Mauldin EA, Morris DO, Goldschmidt MH. Retrospective study: the presence of Malassezia
in feline skin biopsies. A clinicopathological study. Vet Dermatol. 2002;13:7–14.
7. Ordeix L, Galeotti F, Scarampella F, Dedola C, Bardagi M, Romano E, Fondati A. Malassezia
spp. overgrowth in allergic cats. Vet Dermatol. 2007;18:316–23.
8. Yager JA, Gross TL, Shearer D, Rothstein E, Power H, Sinke JD, Kraus H, Gram D, Cowper
E, Foster A, Welle M. Abnormal sebaceous gland differentiation in 10 kittens (‘sebaceous
gland dysplasia’) associated with generalized hypotrichosis and scaling. Vet Dermatol.
2012;23:136–e30.
9. Turek MM. Cutaneous paraneoplastic syndromes in dogs and cats : a review of the literature.
Vet Dermatol. 2003;14:279–96.
10. Gross TL, Clark EG, Hargis AM, Head LL, Hainesh DM. Giant cell dermatosis in FeLV-­
positive cats. Vet Dermatol. 1993;4:117–22.
11. Brachelente C, vonTscharner C, Favrot C, Linek M, Silvia R, Wilhelm S, et al. Non thymoma-­
associated exfoliative dermatitis in 18 cats. Vet Dermatol. 2015;26:40–e13.
12. Dias Pereira P, Faustino AMR. Feline plasma cell pododermatitis: a study of 8 cases. Vet
Dermatol. 2003;14:333–7.
General References
For definitions: Merriam-Webster Medical Dictionary. http://merriam-webster.com Accessed 10
May 2018.
Albanese F. Canine and feline skin cytology. Cham: Springer International Publishing; 2017.
Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, Wolff K. Fitzpatrick’s dermatology in
general medicine. 8th ed. New York: The McGraw-Hill Companies; 2012.
Gross TL, Ihrke PJ, Walder EJ, Affolter VK. Skin diseases of the dog and cat. Clinical and histopathologic diagnosis. 2nd ed. Oxford: Blackwell Publishing; 2005.
Miller WH, Griffin CE, Campbell KL. Muller & Kirk’s small animal dermatology. 7th ed. St.
Louis: Elsevier; 2013.
Noli C, Toma S. Dermatologia del cane e del gatto. 2nd ed. Vermezzo: Poletto Editore; 2011.
VetBooks.ir
Pruritus
Silvia Colombo
Abstract
Pruritus, also called itching, is an irritating sensation in the upper surface of the
skin, thought to result from stimulation of sensory nerve endings. Pruritus is
common in cats and can be further classified based on its distribution (localized
or generalized), location on the animal’s body and severity (mild, moderate, or
severe). From a clinical point of view, pruritus in cats is most commonly caused
by ectoparasitic, allergic, infectious or immune-mediated diseases. Cats manifest
pruritus by overgrooming, which makes it particularly difficult to recognize and
evaluate, and to be differentiated from pain or a behavioral problem. In a very
young cat, ectoparasites and dermatophytosis are common, while in an adult cat,
allergic and immune-mediated skin diseases should also be considered. History
is relevant for concurrent drug administration or systemic disease and for severity and seasonality of pruritus. Most pruritic cats present with one (or more) of
four clinical patterns, namely, head and neck pruritus, miliary dermatitis, self-­
induced alopecia and the eosinophilic granuloma complex. The diagnostic
approach to pruritus should always be carefully followed in each of its steps in
order to make a correct diagnosis.
Definitions
Pruritus, also called itching, is defined as an unpleasant feeling that causes the
desire to scratch. In the vast majority of cases, the irritating sensation develops in
the skin and is thought to result from stimulation of sensory nerve endings. In rare
cases, pruritus may originate in the central nervous system. Pruritus is extremely
common in veterinary dermatology and may be due to a wide variety of diseases.
S. Colombo (*)
Servizi Dermatologici Veterinari, Legnano, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_9
161
VetBooks.ir
162
S. Colombo
It is an obvious clinical sign in dogs, while it can be very subtle in cats because
it can be expressed as excessive grooming, which is a normal feline behavior, or
because cats often hide from owners when they feel the desire to scratch. Pruritus is
further classified based on its distribution (localized or generalized), location on the
­animal’s body and severity (mild, moderate, or severe).
Pathogenesis
The vast majority of the information about mechanisms, pathways, and mediators
of pruritus comes from human or laboratory animal studies and has been reviewed
elsewhere [1, 2]. From a clinical point of view, pruritus in cats is usually caused by
ectoparasitic, allergic, infectious or immune-mediated diseases and can be worsened by concurrent factors such as stress, boredom, dry skin or high environmental
temperature (Table 1). Although pruritus may be interpreted, in some cases, as a
defense mechanism (scratching or licking to remove ectoparasites), skin lesions
often occur as a consequence of behaviors carried out by the cat to relieve it.
Cats manifest pruritus by overgrooming, in other words by increasing the frequency and intensity of a normal, programmed feline behavior. Cats groom to keep
their skin and hair coat clean and healthy, to remove ectoparasites and dirt, to control their body temperature and to relieve tension or stress [3, 4]. Grooming in cats
Table 1 Selected causes of
pruritus in cats
Pruritus
Herpesvirus infection
Superficial pyoderma
Complicated chin acne
Flea infestation
Cheyletiellosis
Notoedric mange
Otodectic mange
Demodicosis (Demodex gatoi)
Trombiculiasis
Dermatophytosis
Malassezia overgrowth
Flea-bite hypersensitivity
Adverse reaction to food
Feline atopic syndrome
Mosquito-bite hypersensitivity
Allergic/irritant contact dermatitis
Adverse drug reaction
Hyperthyroidism
Pemphigus foliaceus
Lymphocytic mural folliculitis
Familial pedal eosinophilic dermatosis
Urticaria pigmentosa-like dermatitis
Idiopathic facial dermatitis of Persian
and Himalayan cats
VetBooks.ir
Pruritus
163
include oral grooming, which is stroking the tongue through the pelage and nibbling
with the incisor teeth, and scratch grooming, which is scratching with the hind paws
[5]. According to one study, indoor, ectoparasite-free adult cats spend 50% of their
time sleeping or resting. Of the time spent awake, oral grooming accounts for about
1 hour per day and scratch grooming for about 1 minute per day. Ninety-one percent
of oral grooming is directed to multiple body regions, while scratch grooming is
always directed to single regions [5].
Being the increased expression of a physiological behavior, overgrooming is
often not recognized by the owner or not interpreted as a sign of pruritus, pain or
stress. Moreover, cats tend to express their discomfort by hiding away from owners,
who may not be aware of their pet’s overgrooming. For all these reasons, pruritus
can be particularly difficult to recognize and evaluate in cats and to be differentiated
from pain (e.g., licking the abdomen due to cystitis) or a behavioral problem (causing licking, scratching, or hair pulling).
Idiopathic ulcerative dermatitis presents as a very severe and extremely pruritic,
usually single, crusted ulceration affecting the dorsal neck (Fig. 1) in which pruritus, neuropathic itch and behavioral disorder have all been considered relevant in
the disease pathogenesis. Idiopathic ulcerative dermatitis is diagnosed by exclusion
of diseases which may induce pruritus to the dorsal neck, such as allergies and
Fig. 1 Idiopathic/
behavioral ulcerative
dermatitis on the dorsal
neck
VetBooks.ir
164
Table 2 Examples of
non-dermatological diseases
to be differentiated from
pruritic skin diseases
S. Colombo
Feline idiopathic cystitis
Psychogenic alopecia
Feline idiopathic/behavioral ulcerative dermatitis
Feline orofacial pain syndrome
Feline hyperesthesia syndrome
Localized neuropathies
ectoparasites. A recent case report proposed that idiopathic ulcerative dermatitis
may be a neuropathic itch syndrome, and the cat responded completely to topiramate, an anti-epileptic drug [6]. However, the same disease has also been investigated from a behavioral point of view. In 13 affected cats, in an open, uncontrolled
study, environmental enrichment and improvement of overall welfare led to resolution of skin lesions, and psychotropic drugs were employed only in one case.
The authors of this study proposed to change the disease name to feline behavioral
ulcerative dermatitis [7].
Finally, an orofacial pain syndrome has been reported in cats. This syndrome
occurs more commonly, although not exclusively, in Burmese cats and is clinically characterized by self-trauma to the face and oral cavity and occasionally by
mutilation of the tongue. The disease may be associated with teeth eruption, dental
disease, and stress and is suspected to be a neuropathic disorder, which should be
considered in cats presenting with severe facial excoriations or ulcers [8].
In conclusion, overgrooming, which includes excessive licking and excessive
scratching, may be the expression of non-dermatological diseases, which should
always be considered when listing the differential diagnoses in an apparently “dermatological” case presenting for “pruritus” (Table 2).
Diagnostic Approach
Signalment and History
Depending on the cat’s age, some diseases may be more likely than others. In a
very young cat, ectoparasites and dermatophytosis are common, especially if the
kitten has been found as a stray or adopted from a cattery, where crowding plays
an important role too. Some diseases, such as dermatophytosis, cheyletiellosis and
notoedric mange, are very contagious. These diseases may affect in contact animals
as well as people, and questions about the presence of skin lesions on other pets
or family members are mandatory. In an adult cat, allergic and immune-mediated
skin diseases should also be considered, while in an older cat, hyperthyroidism may
occur and explain the excessive grooming. Older cats may present with pruritus
due to Malassezia overgrowth, which can be the marker of an underlying systemic
disease or paraneoplastic syndrome (Fig. 2) [9].
VetBooks.ir
Pruritus
165
Fig. 2 Alopecia and
brown greasy material
typical of Malassezia
overgrowth in an old cat
with pancreatic
paraneoplastic alopecia
Fig. 3 Alopecia and
excoriations on the head
and pinnae of a cat affected
by seasonal feline atopic
syndrome
History should include drugs administered for other diseases, which may cause
adverse drug reactions, and ectoparasite prevention. Immunosuppressive therapy or
systemic disease may predispose the cat to dermatophytosis if it gets exposed, for
example, because the owner adopts a new kitten. Seasonality of pruritus may be
useful to limit the list of differentials: ectoparasites and seasonal feline atopic syndrome are more likely in a cat scratching in spring and summer (Fig. 3). Severity of
pruritus should also be analyzed in depth because some diseases are characterized
by extremely severe pruritus (notoedric mange) while in others pruritus may be very
mild (cheyletiellosis, dermatophytosis).
Persian cats of any age are predisposed to dermatophytosis. An older Persian cat
may be affected by dermatophytosis, if it is an asymptomatic carrier, without the
need for contact with a diseased animal [10].
166
S. Colombo
VetBooks.ir
Clinical Presentation
In cats, pruritus is expressed by overgrooming; however, only increased scratching
is easily recognized by the owner. Since they scratch with their hind paws, excoriations usually involve areas that the cat can reach, such as the face, ears, head and
neck. The so called “head and neck pruritus” is a common clinical presentation in
pruritic cats (Fig. 4) [11]. Variably sized excoriations, erosions and ulcers in these
locations may be very severe and deep and are often secondarily infected. This clinical presentation is specifically addressed in Chapter, Excoriations, Erosions and
Ulcers.
Less obviously, alopecia may be caused by an overgrooming, pruritic cat [11].
Self-induced alopecia is characterized by the presence of very short hair fragments
which can be observed by looking closely at the skin or with the help of a magnifying lens. Hair cannot be easily epilated. The alopecic area usually has very well-­
defined margins, with abrupt change to normal hair, and involves parts of the body
that can be reached by the tongue (Fig. 5). Self-induced alopecia is described in
Chapter, Alopecia.
Miliary dermatitis is a peculiar feline clinical presentation also associated with
pruritus [11]. It is characterized by small, crusted papules “resembling millet seeds,”
hence the name, which are more easily felt by touching through the haircoat than
Fig. 4 Excoriations on the
head of an allergic cat
VetBooks.ir
Pruritus
167
Fig. 5 Self-induced
alopecia on the abdomen
of a cat with flea-bite
hypersensitivity
Fig. 6 Small, crusted
papules typical of miliary
dermatitis
seen (Fig. 6). Miliary dermatitis is often associated with self-induced alopecia and
is addressed in Chapter, Papules, Pustules, Furuncles and Crusts.
Another clinical pattern associated with pruritus is a group of lesions named
eosinophilic granuloma complex or eosinophilic dermatitides (Figs. 7 and 8) [12].
These conditions, or clinical presentations, are often caused by allergic diseases and
are discussed in Chapter, Plaques, Nodules and Eosinophilic Granuloma Complex
Lesions.
Many pruritic diseases, in cats, can be associated with one or more of the four
previously described clinical patterns and/or with recurrent otitis (Chapter, Otitis).
However, each disease has its own preferential distribution of pruritus and lesions
on the animal body (Table 3). Some other unusual presentations are also associated
with pruritus and may be caused by hypersensitivity reactions to food or environmental allergens, at least in some cases. Lymphocytic mural folliculitis, for example, is a histopathological reaction pattern occasionally identified in allergic cats
168
S. Colombo
VetBooks.ir
Fig. 7 Eosinophilic
plaques on the abdomen
Fig. 8 Bilateral indolent
ulcer in a domestic
short-haired cat
presenting with pruritus, localized or generalized, partial or complete alopecia and
scaling (Fig. 9) [13]. Urticaria pigmentosa-like dermatitis occurs in Devon Rex or
Sphynx cats and is clinically characterized by an erythematous to hyperpigmented
papular eruption, which is often pruritic (Fig. 10) [14, 15].
Pruritic and non-pruritic dermatoses may be secondarily infected by bacteria or
yeasts. Although this occurs in cats much less frequently than in dogs, one should
always keep into consideration and diagnose/rule out these diseases when examining a pruritic cat [9, 16].
Diagnostic Algorithm
This section is illustrated in Fig. 11. Red squares with numbers represent the steps
of the diagnostic process, as explained below.
Pruritus
169
VetBooks.ir
Table 3 Common locations of selected feline skin diseases associated with pruritus
Locations
Face
Chin
Rump
Thorax, abdomen
Dorsum
Ear canal
Pinnae, paws, abdomen
Pinnae, face, neck, paws, perineum
Head, pinnae, paws, tail, generalized
Chin, claw folds, face, ear canal,
generalized
Rump
Dorsal nose, pinnae, paws
Abdomen, medial thighs, head, neck
Head, pinnae, claw folds, abdomen
Paws
Face
Fig. 9 Alopecia, scaling
and hyperpigmentation on
the head of a cat with
lymphocytic mural
folliculitis
Disease
Herpesvirus infection
Complicated chin acne
Flea infestation
Demodicosis (Demodex gatoi)
Cheyletiellosis
Otodectic mange
Trombiculiasis
Notoedric mange
Dermatophytosis
Malassezia overgrowth
Flea-bite hypersensitivity
Mosquito-bite hypersensitivity
Other allergic diseases
Pemphigus foliaceus
Familial pedal eosinophilic dermatosis
Idiopathic facial dermatitis of Persian and Himalayan cats
VetBooks.ir
170
S. Colombo
Fig. 10 Coalescing,
crusted, and non-crusted
papules in a Sphynx cat
with urticaria pigmentosa-­
like dermatitis
Pruritus
Flea infestation
Demodicosis
Notoedric mange
Otodectic mange
Cheyletiellosis
Trombiculiasis
Skin scraping
Microscopic examination of
hair shafts, skin debris
1
and/or ear cerumen
Wood’s lamp examination
2
Fungal culture
Dermatophytosis
Self-induced alopecia
Malassezia overgrowth
Superficial pyoderma
Complicated chin acne
4 Dentistry examination
Cytology
Histopathology
Eosinophilic inflammation
Head and neck pruritus
Miliary dermatitis
Orofacial pain syndrome
Histopathology
2a
3
Eosinophilic granuloma complex
4
Mosquito-bite hypersensitivity
Familial pedal eosinophilic
dermatosis
Urticaria pigmentosa-like
dermatitis
Urinalysis
Bacterial culture and
sensitivity testing
Ultrasound
Complete blood count
Biochemistry
Endocrinology testing
4
Feline idiopathic cystitis
Hyperthyroidism
Therapeutic trial for fleas
Herpesvirus infection
Idiopathic facial dermatitis of
Persian and Himalayan cats
Pemphigus foliaceus
Adverse drug reaction
Lymphocytic mural folliculitis
Psychogenic alopecia
Idiopathic/behavioral ulcerative dermatitis
Flea-bite hypersensitivity
Behavioral examination
Fig. 11 Diagnostic algorithm of pruritus
6
7
5
Elimination diet
Adverse reaction to food
Feline atopic syndrome
7
Neurologic examination
Feline hyperesthesia syndrome
Localized neuropathies
Pruritus
VetBooks.ir
1
171
Perform skin scrapings and microscopic examination of hair, skin debris and/or
ear cerumen.
In the diagnostic approach to pruritus, it is mandatory to begin with simple
tests to diagnose or rule out ectoparasites. Multiple skin scrapings are useful
for notoedric mange and demodicosis, and fungal spores can be seen surrounding and invading fragments of hair shafts in dermatophytosis. Cheyletiellosis
and trombiculiasis may be diagnosed by microscopic examination of acetate
tape strips, after collecting samples directly from the cat’s coat or, for
Cheyletiella spp., from the material collected from the examination table after
vigorous stroking of the coat. This latter way of collecting specimens may also
be used to find flea dirt, together with coat combing. If pruritus is mainly
affecting the ears, microscopic examination of ear cerumen is required to diagnose otodectic mange.
2 Perform Wood’s lamp examination and fungal culture.
The second step is to rule out or diagnose dermatophytosis, which may
have already been suspected after microscopic examination of hair shafts.
Wood’s lamp examination may support the diagnostic hypothesis and fungal
culture is required to confirm dermatophytosis. If negative results are
obtained, these tests are helpful to rule it out. Since dermatophytosis is common in cats, a fungal culture is appropriate in all cases, although pruritus can
be of variable severity.
2a If the clinical presentation is self-induced alopecia involving the groin and
abdomen, urinalysis, bacterial culture and sensitivity testing, and abdominal
ultrasound should be performed to investigate feline idiopathic cystitis or other
urinary tract diseases. Self-induced alopecia in an old cat may also be caused
by hyperthyroidism, and hematology, biochemistry and endocrine testing
should be carried out in this specific situation.
3 Perform cytology.
Cytology is the easiest and quickest diagnostic test to support the clinical
suspicion of diseases characterized by eosinophilic inflammation, which are
numerous and very common in cats. Eosinophilic plaque and granuloma and
miliary dermatitis are often clinical patterns of allergy, characterized by eosinophilic inflammation, and the diagnostic process should continue to identify the
primary disease. On the other hand, familial pedal eosinophilic dermatosis, mosquito-bite hypersensitivity and urticaria pigmentosa-like dermatitis show eosinophilic inflammation on cytology and are specific diseases which should be
confirmed by histopathological examination. Cytology is also important because
secondary bacterial or yeast infections may complicate the primary disease and
increase the severity of pruritus, although this occurs less frequently in cats compared to dogs. Samples may be taken by impression smear, using a cotton swab
or a piece of acetate tape to look for Malassezia yeasts, bacteria and inflammatory cells. Finally, identification of acantholytic cells admixed with neutrophils
may suggest pemphigus foliaceus.
172
VetBooks.ir
4
5
6
7
S. Colombo
Perform histopathology.
As anticipated, histopathology is the confirmatory diagnostic test for many
feline diseases cytologically characterized by eosinophilic inflammation.
When pruritus affects mainly the face, histopathological examination is
required to diagnose idiopathic facial dermatitis of Persian and Himalayan cats
and herpesvirus infection, although in this latter disease immunohistochemistry may be necessary to confirm the etiology. In cases with clinical manifestation of severe self-trauma to the face and oral cavity, a dental examination may
be required to investigate orofacial pain syndrome. Histopathological examination is useful to diagnose pemphigus foliaceus and, together with history,
adverse drug reactions.
Perform a therapeutic trial for fleas.
In the majority of cases presenting for pruritus, ectoparasites and dermatophytosis can be ruled out at the beginning of the diagnostic approach, and cytological examination only shows secondary infections or eosinophilic
inflammation, which is neither specific nor particularly useful. These cases usually present with one of the four clinical patterns typical of pruritus and should
be investigated in a systematic way. The first step is a therapeutic trial for fleas,
which may have not been identified during the initial investigations for ectoparasites. A positive response to the trial suggests flea-bite hypersensitivity.
Perform an elimination diet.
If the therapeutic trial for fleas is unsuccessful, the second step is performing
an elimination diet with novel protein sources or a hydrolyzed diet, to be carried
out for at least 8 weeks. If the cat improves on the diet, challenge with the previous food is required to diagnose an adverse reaction to food.
After ruling out food as the cause of pruritus, the clinician is left with a possible
diagnosis of feline atopic syndrome. There are different treatment options for
environmental allergy in cats and the diagnosis is confirmed by response to
treatment.
Depending on history and clinical presentation, in some cases, a behavioral
problem can be suspected, especially if the cat presents with self-induced alopecia or ulcerative dermatitis affecting the dorsal neck. In other cases, a neurologic problem such as feline hyperesthesia syndrome may be considered and
needs to be investigated. These conditions are usually addressed only when all
the other differentials have been ruled out, and the cat does not respond to treatment for feline atopic syndrome.
References
1. Metz M, Grundmann S, Stander S. Pruritus: an overview of current concepts. Vet Dermatol.
2011;22:121–31.
2. Gnirs K, Prelaud P. Cutaneous manifestations of neurological diseases: review of neuro-­
pathophysiology and diseases causing pruritus. Vet Dermatol. 2005;16:137–46.
3. Beaver BV. Feline behavior. A guide for veterinarians. Second edition. St. Louis: WB Saunders;
2003.
VetBooks.ir
Pruritus
173
4. Bowen J, Heath S. Behaviour problems in small animals. Practical advice for the veterinary
team. Philadelphia: Elsevier Saunders; 2005.
5. Eckstein RA, Hart BL. The organization and control of grooming in cats. Appl Anim Behav
Sci. 2000;68:131–40.
6. Grant D, Rusbridge C. Topiramate in the management of feline idiopathic ulcerative dermatitis
in a two-year-old cat. Vet Dermatol. 2014;25:226–e60.
7. Titeux E, Gilbert C, Briand A, Cochet-Faivre N. From feline idiopathic ulcerative dermatitis to
feline behavioral ulcerative dermatitis: grooming repetitive behavior indicators of poor welfare
in cats. Front Vet Sci. 2018; https://doi.org/10.3389/fvets.2018.00081.
8. Rusbridge C, Heath S, Gunn-Moore D, Knowler SP, Johnston N, McFadyen AK. Feline
orofacial pain syndrome (FOPS): a retrospective study of 113 cases. J Feline Med Surg.
2010;12:498–508.
9. Mauldin EA, Morris DO, Goldschmidt MH. Retrospective study: the presence of Malassezia
in feline skin biopsies. A clinicopathological study. Vet Dermatol. 2002;13:7–14.
10. Moriello KA, Coyner K, Paterson S, Mignon B. Diagnosis and treatment of dermatophytosis in dogs and cats.: clinical consensus guidelines of the world Association for Veterinary
Dermatology. Vet Dermatol. 2017;28(3):266–8.
11. Hobi S, Linek M, Marignac G, et al. Clinical characteristics and causes of pruritus in
cats: a multicentre study on feline hypersensitivity-associated dermatoses. Vet Dermatol.
2011;22:406–13.
12. Buckley L, Nuttall T. Feline eosinophilic granuloma complex(ITIES): some clinical clarification. J Feline Med Surg. 2012;14:471–81.
13. Rosenberg AS, Scott DW, Erb HN, McDonough SP. Infiltrative lymphocytic mural folliculitis:
a histopathological reaction pattern in skin-biopsy specimens from cats with allergic skin disease. J Feline Med Surg. 2010;12:80–5.
14. Noli C, Colombo S, Abramo F, Scarampella F. Papular eosinophilic/mastocytic dermatitis
(feline urticaria pigmentosa) in Devon rex cats: a distinct disease entity or a histopathological
reaction pattern? Vet Dermatol. 2004;15:253–9.
15. Ngo J, Morren MA, Bodemer C, Heimann M, Fontaine J. Feline maculopapular cutaneous
mastocytosis: a retrospective study of 13 cases and proposal for a new classification. J Feline
Med Surg. https://doi.org/10.1177/1098612X18776141.
16. Yu HW, Vogelnest L. Feline superficial pyoderma: a retrospective study of 52 cases (2001-­
2011). Vet Dermatol. 2012;23:448–e86.
General References
For definitions: Merriam-Webster Medical Dictionary. http://merriam-webster.com Accessed 10
May 2018.
Albanese F. Canine and feline skin cytology. Cham: Springer International Publishing; 2017.
Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, Wolff K. Fitzpatrick’s Dermatology
in General Medicine. 8th ed. New York: The McGraw-Hill Companies; 2012.
Miller WH, Griffin CE, Muller CKL. Kirk’s small animal dermatology. 7th ed. St. Louis: Elsevier;
2013.
Noli C, Toma S. Dermatologia del cane e del gatto. 2nd ed. Vermezzo: Poletto Editore; 2011.
VetBooks.ir
Otitis
Tim Nuttall
Abstract
Otitis externa and media are common in cats, although almost all infections are
secondary. The underlying conditions must be diagnosed and managed for resolution. The approach to feline otitis is different from that in dogs. There are
important differences in the ear anatomy of dogs and cats, although there is less
breed variation among cats. The role of primary, secondary, predisposing and
perpetuating (PSPP) factors is less clear in feline otitis, with fewer predisposing
and perpetuating problems. The primary aetiology of otitis is different from
dogs with less of a role for hypersensitivity dermatoses. There are a variety of
­cat-­specific conditions, including inflammatory polyps, cystoadenomatosis, and
proliferative and necrotising otitis. This chapter will describe the anatomy and
physiology of feline ears, how to use clinical examination, cytology, culture
and imaging in diagnosis, ear cleaning, treatment of otitis externa and otitis
media, and the diagnosis and management of specific ear conditions in cats.
Introduction
Feline otitis requires a different approach to diagnosis and treatment compared to
canine otitis. The aetiology is different and many conditions are specific to cats.
Otitis is less common and less well associated with common skin diseases in cats
than in dogs. For example, otitis has been reported in 16% [1] to 20% [2] of cats
with hypersensitivity dermatitis. In contrast, up to 80% of dogs with atopic dermatitis may suffer from recurrent otitis externa [3]. In addition, the PSPP (primary,
­secondary, predisposing and perpetuating) approach is less useful in cats compared
T. Nuttall (*)
Royal (Dick) School of Veterinary Studies, University of Edinburgh, Roslin, UK
e-mail: tim.nuttall@ed.ac.uk
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_10
175
VetBooks.ir
176
T. Nuttall
to dogs. While it is true that ear infections are invariably secondary and that there
are a number of defined primary causes of otitis in cats, the role of predisposing
factors and perpetuating problems in initiating otitis and the progression to chronic
disease is less clear. Finally, cats can be more sensitive to ototoxicity than dogs, and
topical treatments and ear cleaners must be selected and used with care.
It is very important to recognise that cats with recurrent ear infections have an
underlying problem – they do not have an antimicrobial deficiency! Overuse of antimicrobials can mask the primary condition (which can get more severe and difficult
to manage) and select for antimicrobial resistance (which can complicate future
treatment). Successful management requires diagnosis and appropriate treatment of
the primary cause.
Anatomy and Physiology
Feline ear canal anatomy is similar to that in dogs, although with much less variation among breeds and individuals [4, 5] (Chapter, Structure and Function of the
Skin).
The Pinnae
With the exception of some breeds such as the Scottish Fold, cats have an upright
pinna. The skin of the pinnae and ear canals is continuous with the skin over the rest
of the body. The dorsal surface is covered with densely haired skin that is loosely
attached to the underlying cartilage. The skin on the ventral surface is tightly
attached to the cartilage. Hairs arising on the rostral margin of the pinna fold back
across the pinnal surface and ear canal opening and probably limit entry of foreign
material (Fig. 1). Touching these hairs can elicit ear flicking or head shaking. They
can be extensive in long-haired breeds. The ventral surface of the pinna is otherwise
hairless. There is a complex array of cartilage folds at the base of the pinna. The
most important to recognise is the tragus (Fig. 1), which forms the lateral margin of
the vertical canal. The opening to the vertical ear canal is located behind the tragus.
Ear Canals and Cerumen
The skin of the ear canals is continuous with that of the ventral pinna. It is thin,
hairless and tightly opposed to the underlying ear canal cartilages. The auricular
cartilage is continuous with the pinna and forms the vertical ear canal (Fig. 2). It is
loosely embedded within connective tissue and is reasonably mobile. The auricular
cartilage is connected to the annular cartilage by fibrous tissue, which gives some
flexibility and mobility. The junction is visible as a dorsal ridge extending into the
ear canal lumen (Fig. 3). The annular cartilage forms the short horizontal ear canal
VetBooks.ir
Otitis
177
Fig. 1 The inner surface
of the pinna (rostral is to
the bottom left; caudal to
the upper right). Blue
arrow – hairs arising from
the rostral pinna margin
and extending caudally
across the pinna; black
arrow – the tragus
and is connected to the bony external auditory meatus by fibrous tissue. This gives
some flexibility but the horizontal ear canal is much less mobile than the vertical ear
canal and pinna. The horizontal ear canal is typically 6–9 mm in diameter, which
can limit access of otoscope cones.
Cerumen is typically sparse in healthy cat ears and has a film-like creamy consistency compared to dogs. Outward, lateral migration of the stratum corneum moves
the cerumen, desquamated cells and debris to the opening of the ear canals. Here,
the cerumen dries, detaches and is removed through normal grooming.
Tympanic Membrane
The tympanic membrane (Fig. 4) separates the external ear canals from the middle
ear. It is housed within the bony external auditory meatus (Fig. 2) facing horizontally at an angle from dorso-lateral to ventro-medial, although it can be near vertical
in some cats. The dorsal pars flaccida is narrow and much less prominent than in
VetBooks.ir
178
T. Nuttall
Auricular
cartilage
Annular
cartilage
Auditory ossicles
External
auditory
meatus
Semicircular ducts
Opening of the
Eustachian tube
Cochlea
Dorsolateral
compartment
Tympanic
membrane
Bulla
External ear canal
Promontory
Opening between
bulla compartments
Septum
Ventromedial
compartment of
tympanic cavity
Fig. 2 Schematic diagram of the external ear canals, middle ear and inner ear
Fig. 3 View of the base of
the vertical ear canal. The
arrow indicates the ridge or
shelf formed at the
junction of the vertical and
horizontal ear canals
dogs. The pars tensa forms a thin, grey-white, translucent membrane with prominent striations radiating from the manubrium of the malleus. The malleus forms a
white straight or slightly curved structure running ventrally from the rostro-­dorsal
edge of the tympanic membrane. The concavity of the curve faces rostrally, but this
is less marked than in dogs. The malleus is surrounded by a ring of blood vessels.
VetBooks.ir
Otitis
179
Fig. 4 Normal tympanic
membrane in a healthy cat.
A – attachment of the
malleus; B – pars flaccida;
C – pars tensa
Middle Ear
The middle ear (Fig. 2) is divided by a bony shelf into ventro-medial (pars endotympanica) and dorso-lateral (pars tympanica) compartments. The dorso-lateral compartment is bounded laterally by the tympanic membrane, dorsally by the epitympanic
recess and medially by the lateral wall of the cochlea. The epitympanic recess contains
the auditory ossicles and openings to the cochlear (auditory window) and vestibular
systems (vestibular window). The Eustachian tube opens in the medial wall of the middle ear near the origin of the bony shelf and connects the middle ear with the pharynx.
A sympathetic nerve trunk runs close to the opening of the Eustachian tube. It is quite
superficial in cats and vulnerable when removing polyps, flushing the middle ear and/
or treating otitis (which can result in Horner’s syndrome). The middle ear is lined with
a mucous epithelium continuous with that of the Eustachian tube and pharynx. The
Eustachian tube allows air pressure to equalise across the tympanic membrane and
mucus from the middle ear to drain into the pharynx. A small opening in the bony shelf
allows mucus from the ventral bulla compartment to drain through the Eustachian tube.
General Approach to Otitis
1. Identify and treat the infection.
• Use ear cytology to identify Malassezia, bacteria and inflammatory cells.
• When necessary, perform culture to identify the microorganisms and their
antimicrobial susceptibility.
VetBooks.ir
180
T. Nuttall
2. Identify and manage the primary cause.
• Perform a thorough history and full clinical examination.
• Examine the ear canals for clinical lesions, type of discharge, mites, foreign
bodies, inflammatory polyps and tumours.
• Examine the tympanic membrane for signs of rupture and otitis media.
3. Identify and manage any predisposing and perpetuating causes.
• Clinically assess the extent and severity of chronic inflammatory changes.
• Consider radiographs, CT scans or MRI scans.
Diagnostic Procedures
Examining the Ear
The ears should be carefully examined for any abnormalities. Certain clinical signs
are often highly specific for primary conditions in feline otitis (see below). Healthy
ears should be freely mobile, pliable, non-painful and non-pruritic and have little to no
discharge. Very firm, immobile ear canals are often irreversibly fibrosed and/or mineralised. The skin should be pale, thin and smooth. The ear canals should be open with a
thin, smooth and pale lining, little to no discharge and a normal tympanic membrane.
However, the narrow diameter of cats’ ear canals can make otoscopic examination of
the horizontal ear canal difficult. The tympanic membrane may not be easily visible
in diseased ears. This should not preclude treatment, although the possibility of a ruptured membrane should be considered. Full examination may therefore need sedation,
anaesthesia, removal of any discharge and/or treatment to open the ear canal lumen.
Despite this, careful examination in a conscious cat can help identify the cause of the
otitis as well as the extent and severity of secondary changes without otoscopic examination. The nature of the discharge can indicate the likely problem and/or infection,
but the appearance of the dried discharge at the opening of the ears can be misleading
and fresh material from the ear canals should be evaluated.
Cytology
Cytology is mandatory in all cases of otitis where it can identify the most likely
organisms. This is particularly useful in mixed infections, where culture may identify several organisms with different susceptibility patterns. Samples should be collected from the ear canal with swabs or curettes. Mites can be found in material
collected in mineral oil under x40 magnification. Air-dried or heat-fixed material
stained with a modified Wright–Giemsa stain can be examined under high magnification (x400 or x1000 oil immersion) to see cells and microorganisms.
It is important to recognise biofilms, which form thick, dark and slimy exudates.
They appear as variably thick veil-like material (Fig. 5a, b) on cytology that may
VetBooks.ir
Otitis
181
obscure bacteria and cells. Biofilms are becoming increasingly common in otitis.
Many microorganisms can produce biofilms, which facilitate adherence to the ear
canal epidermis, middle ear lining and surrounding hairs. They also inhibit antimicrobials by providing physical protection and by altering metabolic activity. The net
result is that much higher antimicrobial concentrations predicted by in vitro testing
are required – in effect the minimum inhibitory concentration (MIC) is increased.
Specific anti-biofilm measures (see below) should be used in all cases where they
are present.
The numbers of yeast, cocci, rods, neutrophils and epithelial cells should be
quantified. Staphylococci (Fig. 5b) and Malassezia spp. (Fig. 6) are straightforward
to identify, and a good estimate of their probable sensitivity can be made based on
knowledge of local resistance patterns and previous treatment. Gram-negative bacteria (Fig. 7), however, are harder to differentiate on cytology alone.
Fig. 5 (a) The typical
dark, thick and slimy
appearance of biofilm from
the ear canals; (b) – a
biofilm-associated
staphylococcal infection
showing coccoid bacteria
embedded in a veil-like
matrix (Rapi-Diff II®
stain; ×400 magnification).
Staphylococci typically
form pairs, groups of 4 and
irregular clumps
a
b
VetBooks.ir
182
T. Nuttall
Fig. 6 Malassezia otitis
with large numbers of
budding yeasts (Rapi-Diff
II® stain; ×400
magnification)
Fig. 7 Pseudomonas otitis
with large numbers of rod
bacteria (A) and
neutrophils (B) embedded
in a slimy matrix (C)
consistent with a biofilm.
All bacteria that stain with
modified Wright–Giemsa
stains will stain dark
blue – their Gram-negative
identity can only be
inferred (Rapi-Diff II®
stain; ×400 magnification)
Bacterial Culture and Antimicrobial Susceptibility Testing
Bacterial culture and susceptibility testing is not necessary in most cases of otitis
externa if topical therapy is used, as the antibiotic concentration in these products
greatly exceeds the minimum inhibitory concentration (MIC) for the bacteria.
Great care must be taken in interpreting antibiotic susceptibility and resistance
results because the susceptibility–resistance breakpoints are based on tissue concentrations after systemic treatment. This does not necessarily mean that the bacteria are resistant to the antimicrobial because sufficiently high antibiotic levels, as
achieved with topical therapy, may still exceed the MIC. Sensitivity data are therefore very poorly predictive of the response to topical drugs because concentrations
in the ear canal are much higher. The response to treatment is best assessed using
clinical signs and cytology.
VetBooks.ir
Otitis
183
Bacterial cultures can help identify the bacteria involved in the infection. This
can be useful for less common organisms that are hard to differentiate on cytology
and/or where the antimicrobial susceptibility patterns are less predictable.
Antibiotic sensitivity data should be used to predict the efficacy of systemic
drugs in case these are used (e.g. in case of otitis media), although the concentration
in the ear tissues may be low and high doses are needed. In addition, biofilms that
inhibit antimicrobial penetration and efficacy effectively increase the in vivo MIC,
meaning that in vitro tests over-estimate antimicrobial susceptibility.
Diagnostic Imaging
Diagnostic imaging techniques include radiography, CT and MRI. Radiography
(Fig. 8) is the most widely available but is the least sensitive. A full series should
include dorso-ventral, lateral, right and left lateral oblique, and, where necessary,
rostro-caudal open mouth views [6]. CT scans (Fig. 9) are less widely available,
but are fast, can be done under sedation, and are highly accurate for bony and softtissue changes. Post-contrast evaluation of bone- and soft-tissue weighted views can
reveal the extent and severity of inflammation as well differentiate tissue density
Fig. 8 Rostro-caudal open
mouth radiograph of a cat
with unilateral otitis media.
The left middle ear
compartments have a
normal dark air-filled
appearance. The right
middle ear is filled with
opaque material consistent
with soft-tissue or fluid
T. Nuttall
VetBooks.ir
184
Fig. 9 CT scan of a cat with bilateral otitis media. There is some thickening of the tympanic bulla
wall consistent with chronic inflammation (compare to the bulla walls in Fig. 8). The ventral compartments are filled with soft tissue, which density analysis revealed to be fluid. The right dorsal
compartment is also filled with soft tissue that was shown to be solid. The cat had an inflammatory
polyp in the right ear and mucoid congestion in the ventral bullae in both ears
(e.g. solid tissue, fat and fluid). MRI is best for evaluating the soft tissues and nerves
around the ears, but will not image bony structures adequately.
Ear Cleaning and Ear Flushing
Ear cleaning removes debris and microbes from the canal [4, 5, 7]. Some ear cleaners have broad-spectrum antimicrobial activity [8]. Very waxy or exudative ears
should be cleaned daily during treatment, but this isn’t necessary if there is less
debris. It is important to demonstrate effective ear-cleaning techniques to owners.
Ear Cleaners
Ceruminolytic (lift debris off the epidermis) and ceruminosolvent (soften cerumen)
cleaners (i.e. propylene glycol, lanolin, glycerine, squalane, butylated hydroxytoluene, cocamidopropyl betaine and mineral oils) are useful for softening and removing dry waxy debris and/or wax plugs. Surfactant-based ear cleaners (i.e. docusate
sodium, calcium sulfosuccinate and similar detergents) are better in more seborrhoeic ears and purulent ears. Tris-EDTA has very little ceruminolytic or detergent activity, but is soothing in ulcerated purulent ears and is safer if the tympanic
VetBooks.ir
Otitis
185
membrane is ruptured. Astringents (i.e. isopropyl alcohol, boric acid, benzoic acid,
salicylic acid, sulphur, aluminium acetate, acetic acid and silicon dioxide) can
help prevent maceration of the epithelial lining of the canal. Antimicrobials (e.g.
p-­chlorometaxylenol [PCMX], chlorhexidine and ketoconazole) can help treat and
prevent infections. Tris-EDTA has little antimicrobial activity by itself, but high
concentrations can potentiate the effect of antibiotics and chlorhexidine [9, 10]. Ear
cleaners should be used with some caution in cats, and some ingredients (e.g. detergents, acids and alcohols) can irritate the ears and/or trigger ototoxicity.
Ear Flushing
Thorough ear flushing under general anaesthetic is the only way to clean the deeper
ear canal and middle ear [4, 5, 7]. A tomcat catheter, urinary catheter or feeding tube
can be inserted into the ear canals and, if necessary, middle ear under direct visualisation through an operating head or, preferably, video otoscope. The longer length,
narrower diameter, magnification and visual clarity of a video otoscope make the
procedure much easier, more accurate and safer. Ear flushing should be done with
saline or water to minimise the risk of ototoxicity or Horner’s syndrome. This is
flushed and aspirated into the ear canals and/or middle ear until clean. It may be
necessary to use a ceruminolytic initially to help soften and loosen stubborn debris,
but this should be carefully and thoroughly flushed out afterwards.
Myringotomy
A myringotomy (deliberate rupture of the tympanic membrane) should be considered if the ear drum is intact but there is evidence of middle ear disease (e.g. clinical
signs, abnormal tympanic membrane and/or diagnostic imaging findings) [4, 5, 7].
A catheter, stylet, spinal needle or curette can be used to puncture the ventro-lateral
portion of the tympanic membrane (Fig. 4). This avoids the important structures in
the epitympanic recess. It’s only possible to get access to the dorso-lateral tympanic
bulla through the tympanic membrane as the bony shelf (while allowing some communication) prevents direct entry into the ventro-medial compartment.
Diseases of the Pinna
Diseases of the pinna (Table 1) are relatively common in cats. They are normally
associated with more generalised skin conditions that are covered in detail elsewhere in this book. In contrast to the more specific causes of otitis, these conditions
rarely affect the ear canals.
186
T. Nuttall
VetBooks.ir
Table 1 Diseases of the pinna
Alopecia
Pruritic and eosinophilic dermatitis
Pustules and crusting
Necrosis and ulceration
Thickening, scaling and pigmentation (with
or without distortion)
Nodules and ulcers
Nodules, ulcers and sinus tracts
Hyperadrenocorticism
Hypothyroidism (very rare)
Dermatophytosis
Demodex spp. (rare)
Follicular dysplasia (Devon rex cats)
Lymphocytic and other mural folliculitis
Alopecia mucinosa
Mosquito-bite hypersensitivity
Other biting insects (including rabbit fleas)
Head and neck dermatitis
Eosinophilic granuloma complex
Notoedres cati and Sarcoptes scabiei (rare)
Pemphigus foliaceus
Drug reactions
Vasculitis
Cold agglutinin disease
Frostbite
Actinic keratosis
Cutaneous horn
Multicentric squamous cell carcinoma in situ
(Bowen’s disease)
Auricular chondritis
Squamous cell carcinoma
Cat bite abscess or cellulitis
Aural haematoma (rare)
Cryptococcus spp. and other deep fungal
infections
Deep bacterial infections (e.g. Actinomyces and
Nocardia spp.)
Mycobacterial infections
Otitis Externa
Clinical Signs
Clinical signs of otitis externa include pruritus, head shaking, inflammation and
discharge. The division into erythroceruminous and suppurative otitis is less clear
than in dogs. Waxy ceruminous to seborrheic discharges may be sterile or involve
Malassezia or (less commonly) a bacterial overgrowth (i.e. there are no neutrophils
or other inflammatory exudates). Purulent otitis is relatively more common than in
dogs (where erythroceruminous otitis predominates). However, severe ulcerative
Pseudomonas otitis is uncommon in cats.
Acute, unilateral otitis externa can be seen with foreign bodies, whereas chronic
unilateral otitis suggests neoplasia or an inflammatory polyp. Bilateral otitis externa
in cats is most commonly associated with otodectic mange, but chronic cases can be
associated with adverse food reactions or other hypersensitivity diseases.
Otitis
187
VetBooks.ir
Table 2 Primary, predisposing and perpetuating factors in otitis externa
Primary (the actual cause
of the ear disease)
Predisposing (make otitis
more likely or more likely
to be severe)
Perpetuating (prevent
resolution)
Otodectes cynotis
Demodex spp. (rare)
Foreign bodies (rare)
Adverse food reactions
Feline atopic syndrome (feline atopic dermatitis)
Inflammatory polyps
Cystoadenomatosis
Proliferative and necrotising otitis
Ceruminous gland neoplasia
Seborrheic otitis
Conformation (e.g. pendulous, hairy, narrow ears and
ceruminous ears; rare in cats)
Swimming (very rare in cats)
Maceration or irritation of canal epithelium with cleaning or
medication
Chronic pathological changes (e.g. decreased epithelial
migration, sebaceous and ceruminous hyperplasia, increased
discharge, oedema, fibrosis, thickening and stenosis, and
calcification)
Otitis media
The PSPP Approach
Ear infections are almost always secondary to primary, predisposing and perpetuating factors (the PSPP approach – Table 2) [4, 5]. The primary cause of the otitis
must be diagnosed and treated. Predisposing factors may not be easily countered or
managed, but should alert clinicians to animals that will be more likely than others
to have recurrent otitis. However, with the exception of excessive cleaning or medication, these are less important in cats compared to dogs. Failure to address all the
perpetuating causes of otitis externa commonly results in relapsing chronic otitis.
Perpetuating causes can change over the course of chronic otitis and will eventually
lead to irreversible changes that require a total ear canal ablation.
Bacterial and Malassezia Infections
Staphylococcal bacteria (e.g. Staphylococcus pseudintermedius and S. felis) are
most common, but other organisms can include streptococci, Pasteurella multocida,
E. coli, Klebsiella pneumoniae, and/or Proteus, Pseudomonas, Corynebacteria and
Actinomyces spp. [4, 5]. Mixed bacterial infections are frequently seen. Many staphylococci and Gram-negative strains can produce biofilms [11]. These inhibit cleaning, prevent penetration and activity of antimicrobials (effectively increasing the
MIC) and provide a protected reservoir of bacteria (Figs. 5 and 7). They may also
enhance the development of antimicrobial resistance, especially in Gram-­negative
bacteria that acquire stepwise resistance mutations to concentration-­
dependent
antibiotics.
188
T. Nuttall
VetBooks.ir
Table 3 First-line topical antimicrobials
Fusidic acid
Florfenicol
Polymyxin B
Gentamicin
Neomycin
Fluoroquinolones
Nystatin
Terbinafine
Clotrimazole
Miconazole
Posaconazole
Gram-positive only
Effective against MRSA and MRSP
Synergistic with framycetin against Gram-positive bacteria
Broad-spectrum but not effective against Pseudomonas spp.
Broad-spectrum and effective against Pseudomonas spp.
Inactivated by organic debris and needs a clean ear canal
Synergistic with miconazole against Gram-negative bacteria
Broad-spectrum and effective against Pseudomonas spp.
Broad-spectrum but limited efficacy against Pseudomonas spp.
Broad-spectrum and effective against Pseudomonas spp.
Additive activity with silver sulfadiazine against Pseudomonas spp.
Broad-spectrum antifungals
MRSA methicillin-resistant Staphylococcus aureus, MRSP methicillin-resistant S. pseudintermedius
First-Line Antimicrobial Treatment
In general, topical antimicrobials are more effective than oral antibiotics for resolving otitis externa (Table 3). High antimicrobial concentrations (usually mg/ml) can
overcome apparent antibiotic resistance. It is important to use an adequate volume
to penetrate into the ear canals – 0.5–1 ml is sufficient for most cats, but this may be
too much in very small animals.
The efficacy of concentration-dependent drugs (e.g. fluoroquinolones and aminoglycosides) depends on delivering concentrations of at least 10x MIC once daily.
Time-dependent drugs (penicillins and cephalosporins) require concentrations
above MIC for at least 70% of the dosing interval. This is readily obtained with
topical therapy, which achieves high local concentrations that probably persist in
the absence of systemic metabolism. Most topical medication should be effective
with once daily dosing, although some are licensed for twice daily administration.
Products licensed for use in dogs should be used with care in cats as ototoxicity is
possible.
Topical Antibiotic Treatment of Multi-drug-Resistant Bacteria
If bacteria persist on cytology despite 1–2 weeks of appropriate treatment, then antibiotic resistance should be suspected. Other reasons for treatment failure include
polyps, neoplasia, foreign bodies and other underlying conditions; debris, biofilms
and failure to clean the ears; stenosis, otitis media and other perpetuating factors;
and poor compliance. Pseudomonas are inherently resistant to many antibiotics, and
they readily develop further resistance if treatment is ineffective. There are a variety
of approaches to multi-drug-resistant infections (Table 4), although none of these
are licensed in cats and they must be used with care.
Otitis
189
Table 4 Antibiotics useful in multi-drug-resistant infections
VetBooks.ir
Antibiotic
Ciprofloxacin
Enrofloxacin
Marbofloxacin
Clavulanate–
ticarcillina
Ceftazidimea
Silver
sulfadiazine
Amikacin
Gentamicin
Treatment regimes
0.2% sol. 0.15–0.3 ml/ear q 24 h
2.5% injectable sol. diluted 1:4 with TrizEDTA, saline or Epi-Otic®
topically q 24h; 22.7 mg/ml sol. 0.15–0.3 ml/ear q 24 h
1% injectable sol. diluted 1:4 with saline or TrizEDTA topically q 24h;
2 mg/ml in TrizEDTA topically q 24h; 20 mg/ml sol. 0.15–0.3 ml/ear q 24h
16 mg/ml in TrizEDTA topically q 24h; reconstituted injectable sol.
0.15–0.3 ml/ear q 12h; 160 mg/ml sol. 0.15–0.3 ml/ear q 12h; potentially
ototoxic
10 mg/ml in TrizEDTA topically q 24h; 100 mg/ml 0.15–0.3 ml/ear q 12h
Dilute to 0.1–0.5% in saline; additive activity with gentamicin and
fluoroquinolones
2 mg/ml in TrizEDTA topically q 24h; 50 mg/ml 0.15–0.3 ml/ear q 24h;
susceptibility maintained if there is resistance to other aminoglycosides;
potentially ototoxic
3.2 mg/ml in TrizEDTA topically q 24; ototoxicity possible but uncommon
Reconstituted sol. Stable for up to 7 days at 4 °C or for 1 month frozen
a
Tris-EDTA
Tris-EDTA damages bacterial cell walls and increases antibiotic efficacy, which
can overcome partial resistance. It is best given 20–30 minutes before the antibiotic
but can be co-administered. It is well tolerated and non-ototoxic. Tris-EDTA shows
additive activity with chlorhexidine, gentamicin and fluoroquinolones at high concentrations [9, 10, 12].
Treatment of Biofilms and Mucus
Biofilms can be physically broken up and removed by thorough flushing and aspiration. Topical Tris-EDTA and N-acetylcysteine (NAC) can disrupt biofilms, facilitating their removal, and enhancing penetration of antimicrobials. NAC, however,
is potentially irritating (particularly in concentrations above 2%). Systemic NAC
(600 mg orally twice daily) can help dissolve biofilms in the middle ear. Systemic
NAC and bromhexine (1–2 mg/kg orally q12h) can liquefy mucus, facilitating
drainage in otitis media due to chronic mucosal inflammation of inflammatory polyps (see below).
Systemic Antimicrobial Therapy
Systemic therapy may be less effective in otitis externa because bacteria are present only in the external ear canal and cerumen, there is no inflammatory discharge
and penetration to the lumen is poor. Systemic treatment is indicated when the ear
canal cannot be treated topically (e.g. stenosis or compliance problems or if topical
VetBooks.ir
190
T. Nuttall
adverse reactions are suspected) and in otitis media. High doses of drugs with good
tissue penetration (e.g. clindamycin or fluoroquinolones) should be considered.
Oral itraconazole (5 mg/kg orally once daily) can be administered if systemic antifungal therapy is indicated.
Otitis Media
Aetiology and Pathogenesis
Otitis media may be primary or secondary. Inflammatory polyps (see below) are the
most common cause of primary otitis media in cats. Chronic otitis externa can result
in maceration and rupture of the tympanic membrane (Fig. 10), especially with
stenosis of the horizontal ear canal and Gram-negative bacterial infections. Chronic
upper respiratory infections lead to inflammation and increased bacterial colonisation of the nasopharynx, which may ascend up the Eustachian tube. Less commonly,
infections may spread to the middle ear from retrobulbar or para-aural abscesses, or
other severe local or systemic infections. Middle-ear infections may rupture through
the tympanic membrane into the ear canal or spread to the para-aural tissues and/or
central nervous system.
Clinical Signs
Clinical signs associated with otitis media can be chronic, mild and vague until
neurological deficits become evident (Table 5) (Figs. 11 and 12).
Fig. 10 Ruptured ear drum
in cat with chronic upper
respiratory tract and middle
ear infections. The middle
ear mucosa (A) is visible
behind the malleus (B)
Otitis
191
VetBooks.ir
Table 5 Clinical signs associated with otitis media in cats
Otitis media
Head shaking, rubbing or scratching
Dullness and pain; may avoid hard
foods and handling around the head
Reduced ability to localise sound
(unilateral)
Deafness (bilateral)
Neurological deficits
Horner’s syndrome (miosis, partial ptosis and
apparent enophthalmos); sympathetic trunk
Ataxia and nystagmus; peripheral vestibular
syndrome
Facial paresis; facial nerve
Fig. 11 Head tilt in a cat
with otitis media
associated with a
Pasteurella infection
Diagnosis
Using otoscopy and myringotomy to demonstrate an abnormal tympanic membrane
and/or fluid or debris in the tympanic bulla is diagnostic. Imaging such as radiography, CT or MRI is useful when stenosis limits otoscopic examination and will
reveal the extent of the otitis media and lytic, proliferative and/or expansive changes
in the tympanic bulla.
VetBooks.ir
192
T. Nuttall
Fig. 12 Horner’s
syndrome in a cat with
otitis media caused by an
inflammatory polyp
Treatment
Many cases of otitis media will resolve with medical therapy, but most cases will
also require flushing of the middle ear. A myringotomy will be necessary if the
tympanic membrane is intact.
Systemic antibiotics should be selected based on bacterial culture of the middle ear, taking into account the ability of the antibiotic to penetrate the middle
ear. Penetration of antibiotics into chronically inflamed ears could be poor, and
high doses of drugs with a high volume of distribution (e.g. fluoroquinolones)
should be considered [13]. Systemic therapy can be challenging if susceptibility
is limited to topical and/or parenteral drugs. Topical antibiotics and glucocorticoids in saline or Tris-EDTA can be directly administered into the middle ear;
gentamicin, fluoroquinolones, ceftazidime and dexamethasone do not appear to
be specifically ototoxic when administered this way [5, 14]. It is unclear how
long these drugs persist in the middle ear, but, as this is essentially a blind-ended
sac, drugs are likely to be active in the middle ear for a few days. Repeated
instillation of mg/ml solutions every 5–7 days could therefore be useful in multidrug-resistant infections that are refractory to oral medication. Treatment should
continue until clinical resolution, which may take 6–8 weeks in severe cases.
Oral or topical glucocorticoids and/or mucolytics should also be administered in
appropriate cases. The tympanic membrane generally heals within 2–3 weeks if
infection and inflammation are controlled.
Chronic otitis media, osteomyelitis of the tympanic bulla, cholesteatoma and
para-aural abscesses may be refractory to medical treatment. A total ear canal ablation/lateral bulla osteotomy is required in these cases.
Otitis
193
VetBooks.ir
Foreign Bodies
Clinical Signs
Potential foreign bodies include grass awns, hair and other organic debris, cotton
wool, cotton bud tips and (occasionally) broken pieces from catheters or forceps.
These cause variable, acute and sometimes extreme pain and pruritus. Some cases
may present with chronic otitis externa that is poorly responsive to treatment. Most
cases are unilateral but bilateral foreign bodies can be seen. Foreign bodies may
penetrate the tympanic membrane and cause otitis media.
Diagnosis
Otoscopic examination will reveal variable amounts of inflammation and exudates. The nature of the exudate will depend on the secondary infection. Large
foreign bodies are usually visible, but the cat may need to be sedated or anaesthetised to allow ear cleaning and complete visualisation of the ear canal. Advanced
imaging may be needed to detect a foreign body that has penetrated fully into the
middle ear.
Treatment
The foreign body must be removed using forceps and/or ear flushing. Care should
be taken to check that it has been completely removed – tiny fragments left behind
can be enough to perpetuate the problem. The inflammation and secondary infection
should be treated appropriately (see above), but they normally quickly resolve once
the foreign body has been removed.
Inflammatory Polyps
Aetiology and Pathogenesis
Inflammatory polyps are a common cause of otitis media and externa in cats
[15]. They are most common in young cats but can be seen in older individuals.
Most are unilateral but they can be bilateral. Their aetiology is unknown but may
involve an abnormal inflammatory reaction to the commensal nasopharyngeal
microflora or to respiratory virus infections (although virus isolation has been
negative) [16].
194
T. Nuttall
VetBooks.ir
Clinical Signs
The polyps usually arise in or near to the opening of the Eustachian tube [15]. They
may extend down the Eustachian tube and into the nasopharynx, causing snoring,
an altered voice, sneezing, coughing, gagging and/or retching. Polyps within the ear
usually present with otitis media (if the tympanic membrane is intact; Fig. 12) and/
or otitis externa (if the tympanic membrane is ruptured and the polyp extends into
the ear canals). This is usually associated with a secondary bacterial infection and
a purulent discharge in the ear canals (Fig. 13). It is unusual for the polyp to extend
in the ventro-medial compartment of the tympanic bulla. However, this is often full
of mucus arising from obstruction of the Eustachian tube and draining foramen by
the polyp and inflamed mucosa (Fig. 9). This can become stagnant, inspissated and/
or infected.
Diagnosis
A history of unilateral otitis, purulent discharge, Horner’s syndrome and/or otitis
media is highly suggestive of an inflammatory polyp [15]. The polyp may be visible
in the ear canal (after cleaning any discharge; Fig. 14) or in the nasopharynx (which
usually requires sedation and pulling the soft palate rostrally). Diagnostic imaging
can be used to confirm the extent and severity of the polyp and secondary infection.
Computerized tomography (CT) has much better sensitivity and specificity than
radiography. Density analysis of the pre- and post-contrast CT images weighted
for bone and soft tissue will differentiate solid polyp tissue from fluid (mucus and/
or pus) and show areas of active inflammation or infection (Fig. 9). This allows
Fig. 13 Waxy and
suppurative debris in the
ear canal of a cat with an
inflammatory polyp. The
polyp could not be seen
until the debris was flushed
away. Repeated courses of
antibiotics had led to an
MRSA infection
VetBooks.ir
Otitis
195
Fig. 14 Inflammatory
polyp in the horizontal ear
canal
accurate assessment of the extent of the polyp; for example, while radiographs can
suggest that all compartments of the middle ear are affected, a CT scan can show
the true extent of the solid polyp, mucus build-up and involvement of the Eustachian
tube. CT scans will also more accurately assess changes to the bony structures of
the tympanic bulla and middle ear (e.g. osteomyelitis, sclerosis, proliferation and/
or lysis). This can be critical for planning treatment; for example, in most cases, CT
scans show that solid polyps don’t extend into the ventral compartment, making a
ventral bulla osteotomy unnecessary. Other types of tumours should be suspected if
a polyp-like mass develops in older cats, and histopathology can be used to confirm
the diagnosis.
Treatment
Polyps in the nasopharynx, dorsolateral middle ear and external ear canal can be
removed via traction with forceps under anaesthetic [17, 18]. The polyp is grasped
firmly with a set of forceps and gradual continuous traction applied to pull the polyp
from the middle ear. They can usually be removed in one piece, but sometimes multiple attempts are needed (Fig. 15). Twisting the polyp stalk before or during traction
can help limit bleeding. Small polyps in the horizontal ear canal can be removed
using alligator forceps through an operating head or video otoscope. Flatter nodules
that are difficult to grasp with forceps can be ablated with a laser (Fig. 16). Solid
polyps in the ventro-medial bulla compartment have to be removed via a ventral
bulla osteotomy, although this is uncommon.
Otitis externa and media are often present, and material from the ear canals and
middle ear should be collected for cytology and culture. The polyps themselves
VetBooks.ir
196
T. Nuttall
are invariably sterile. The ear canals and middle ear should be flushed and treated
appropriately (see treatment of otitis externa and otitis media above).
Systemic glucocorticoids (e.g. 2 mg/kg prednisolone or 0.2 mg/kg of dexamethasone daily to resolution and then slowly tapered) will reduce post-traction
inflammation, help open the bulla foramen and Eustachian tube and may help to
reduce the recurrence rate [19]. N-acetylcysteine (600 mg orally q12h) or bromhexine (2 mg/kg q12h) can help liquefy mucus and facilitate drainage from the
middle ear.
Fig. 15 Inflammatory
polyp (see Fig. 14) after
removal by traction. The
intact stalk indicates that it
has been successfully
removed intact
Fig. 16 (a) Sessile
inflammatory polyps
arising in the horizontal ear
canal; (b) the horizontal
ear canal following laser
ablation of the polyps and
ear flushing
a
Otitis
b
VetBooks.ir
Fig. 16 (continued)
197
Potential complications include Horner’s syndrome, vestibular syndrome and
facial nerve paralysis. Horner’s syndrome is particularly common as a sympathetic
nerve trunk is very close to the most common origin of polyps near the opening of
the Eustachian tube (Fig. 12). These problems are usually temporary, and permanent deficits are rare.
Cystoadenomatosis (Cystomatosis)
Aetiology and Pathogenesis
Cystoadenomatosis results in multiple, pigmented, ceruminous papules, nodules and
cysts in the ventral pinna and external ear canal [4, 5, 20]. The cause is unknown,
but may involve genetic predisposition and inflammatory triggers. Persian cats may
be predisposed. The cysts eventually block the ear canal resulting in otitis externa
and secondary infections.
Clinical Signs
The clinical appearance is highly suggestive. Early cases present with multiple
blue-black comedones and papules around the opening of the ear canal (Fig. 17).
These gradually increase in number and size, forming nodules and cysts that may
spread onto the pinna and into the vertical and (less commonly) horizontal ear
canals (Fig. 18). Ruptured cysts release a brown-black fluid. Cysto adenomatosis is
VetBooks.ir
198
Fig. 17 Multiple
blue-black comedones and
cysts on the pinna of a cat
with cysto adenomatosis
Fig. 18 Multiple cysts
obstructing the ear
canals in a cat with
cysto adenomatosis
T. Nuttall
VetBooks.ir
Otitis
199
not usually pruritic or painful unless secondary infections are present. Ceruminous
adenomas and adenocarcinomas are more commonly seen in older animals and
form solitary or small numbers of discreet tumours.
Diagnosis
The clinical appearance is largely pathognomonic. If necessary, histopathology will
differentiate adenocarcinoma, adenoma and cysto adenomatosis. Ear cytology can
be used to identify secondary Malassezia and/or bacterial infections. Advanced
imaging can be performed if an otitis media is suspected.
Treatment
Medical treatment is suitable for early and/or mild cases. Any secondary otitis should be treated appropriately. Systemic and/or topical steroids can decrease
swelling and ceruminous hyperplasia in the ears. Regular maintenance therapy with
topical steroids can be used to maintain remission.
More extensive nodules and cysts can be ablated with a CO2, diode or other laser,
or electrocautery (Fig. 19). Topical antibiotics/steroid combinations should be used
postoperatively to reduce inflammation and prevent infection. Laser ablation is very
effective with prolonged remission. Regular topical steroids may reduce the recurrence rate.
Fig. 19 Same cat as in
Fig. 18; here the cysts have
been ablated using a laser
VetBooks.ir
200
T. Nuttall
Total ear canal ablation with lateral bulla osteotomy is necessary in cases where
medical therapy or laser ablation isn’t appropriate or available. Surgery is curative,
although at the expense of the ear.
Otodectes and Other Parasites
Aetiology and Pathogenesis
Otodectes cynotis mites are the most common cause of parasitic otitis, although
others can include trombiculid mites (mainly on the pinnae), Demodex species (mainly on the pinnae and only rarely in the ear canals) and Otobius megnini (the spinose ear tick; very rare in cats). Otodectes are highly contagious
between cats and other species, and are common in multi-cat situations (particularly with young animals and/or where there is a high turnover). Most Demodex
are not contagious, although Demodex gatoi may be contagious between cats in
a household.
Clinical Signs
Otodectes cynotis infestation is characterised by large amounts of dry, brown, waxy
debris with variable erythema and pruritus (Fig. 20). The pruritus may be severe
and extend onto the head and neck, making Otodectes a differential diagnosis in
head and neck dermatitis. Cats can develop hypersensitivity reactions to Otodectes,
and small numbers of mites can still be associated with clinical signs. Some cats
are apparently asymptomatic carriers of Otodectes. Demodex species cause similar
clinical signs. Otobius can result in a variably severe inflamed and painful otitis
externa.
Diagnosis
The history and clinical signs are highly suggestive of Otodectes. Careful otoscopic
examination often reveals the mites as they move (Fig. 21). Otodectes and Demodex
can be seen on microscopic examination of the waxy debris broken up in liquid
paraffin (Fig. 22). Treatment trials for Otodectes (see below) are warranted if mites
aren’t found as small numbers can be missed in sensitised cats. Demodex may be
found on hair plucks, tape strips or skin scrapes of the pinnae or elsewhere. Otobius
ticks should be obvious on otoscopic examination.
VetBooks.ir
Otitis
Fig. 20 Characteristic dry
waxy discharge at the
opening of the ear canal in
a Persian cat with
Otodectes
Fig. 21 Otodectes seen in
the ear canal using a video
otoscope
201
VetBooks.ir
202
T. Nuttall
Fig. 22 Adult Otodectes
mite found by collecting
the ear canal discharge into
liquid paraffin (x100
magnification)
Treatment
Most anti-mite products are effective against Otodectes, including topical fipronil,
selamectin and imidacloprid/moxidectin. Isoxazoline drugs seem to be highly effective against Otodectes and Demodex. All potentially exposed cats and dogs should
be treated. Otobius ticks can be killed with an appropriate product before careful
removal using forceps. Any secondary otitis usually resolves rapidly but can treated
if necessary.
Neoplasia
Aetiology and Pathogenesis
Ear canal neoplasia is fairly common in older cats. Most tumours are ceruminous
adenomas, but malignant adenocarcinomas comprise up to 50% of ceruminous
tumours [4, 5, 20]. Other tumours in the ear canals are rare. The obstruction usually
results in a secondary otitis externa. Malignant tumours can result in local destruction and invasion and potentially spread to local lymph nodes and distal organs.
Tumours arising from external tissues can occasionally invade the middle ear and/
or ear canals.
Otitis
203
VetBooks.ir
Clinical Signs
Most tumours arise at the base of the pinna and upper vertical canal, although they
can be seen at any depth in the ear canals. The nodules can be obscured by discharge
if there is a secondary infection. Swelling of the surrounding tissues and/or local
lymph nodes is suggestive of local spread and/or metastasis.
Diagnosis
The clinical presentation is usually obvious, although single lesions deeper in the
ear canal should be distinguished from inflammatory polyps and multiple tumours
from cysto adenomatosis. Cytology and/or histopathology can help differentiate
neoplasia from polyps and benign from more malignant tumours (Fig. 23). Aspirates
should be taken from local lymph nodes if there is any suspicion of malignancy
or spread. Imaging (especially CT scans) can be used to determine the extent and
severity of local invasion, lymph node involvement and metastasis to internal organs
such as the lungs.
Treatment
Accessible benign tumours can be surgically excised. Lasers can be used to excise
and ablate tumours deeper in the ear canals where surgical excision isn’t possible.
Vertical ear canal surgery or total ear canal ablation can be used to remove tumours
if lasers aren’t available or wider surgical margins are required (Fig. 24). The prognosis is good unless there has been metastatic spread.
Fig. 23 Fine needle
aspiration cytology from a
ceruminous adenoma;
there are numerous
epithelial cells forming a
well-defined and
differentiated sheet with
minimal pleomorphism
VetBooks.ir
204
T. Nuttall
Fig. 24 Benign
ceruminous adenoma in
the horizontal ear canal of
a cat following a total ear
canal ablation. A laser
could have been used to
ablate the tumour in situ
and preserve the ear canal
Seborrheic Otitis
Seborrheic otitis is a common problem of uncertain aetiology and clinical significance [4, 5]. Dark waxy to greasy scales build up on the inside of the pinna and
around the opening of the ear canal (Fig. 25). The lower vertical and horizontal ear
canals are usually normal. The cats may have no other clinical signs but can shake
their heads, flick their ears or scratch at their ears.
This may be secondary to inflammation, and affected cats should be carefully
evaluated for other clinical signs consistent with primary causes of otitis, which
should be managed appropriately. Seborrheic otitis may be seen in Persian and
related cats with idiopathic facial dermatitis. Sphynx and Devon rex cats can have
VetBooks.ir
Otitis
205
Fig. 25 Build-up of
sebaceous/ceruminous
material in the vertical ear
canal. The focal
accumulation of the
material is suggestive of
glandular hyperplasia or
hypersecretion
an asymptomatic build-up of cerumen on the inner pinnae. Cytology should be used
to determine whether there is a bacterial or Malassezia infection. However, the
waxy build-up may simply be material accumulating on the pinna after epidermal
migration out of the ear canals. If there are no other clinical signs, the cats don’t
need treatment. If necessary, gentle wiping with a ceruminosolvent/ceruminolytic
ear cleaner can reduce the build-up.
Proliferative and Necrotising Otitis
Aetiology and Pathogenesis
The aetiology of this condition is unknown, but it is probably immune-mediated [21,
22]. The lesions show T-cell-mediated keratinocyte apoptosis similar to erythema
multiforme [22]. It was first recognised in kittens but has now been reported in adult
and older cats, although most cases are seen in cats less than 4 years old [4, 5].
Clinical Signs
The clinical signs are very suggestive. The lesions are usually symmetrical and most
commonly affect the base of the pinna, opening of the ears and the vertical ear canal.
Occasionally, the lips, face, periocular skin or remote sites may become involved.
Affected cats develop erythematous and hyperkeratotic plaques with tightly adherent
crusts. More severe cases may have erosions, ulceration and haemorrhage (Fig. 26).
Secondary bacterial or Malassezia ear infections may obscure the clinical lesions.
VetBooks.ir
206
T. Nuttall
Fig. 26 Proliferative and
necrotising otitis in a cat
with erythematous plaques,
erosions and crusts
Diagnosis
The diagnosis is usually based on the history and clinical signs. Cytology should
be used to identify secondary infection, which should be treated appropriately [21].
Where necessary, cytology and histopathology can be used to confirm the diagnosis and eliminate differential diagnoses affecting the pinna and ear canals such as
pemphigus foliaceus, eosinophilic granuloma syndrome and thiamazole-associated
drug reactions.
Treatment
The prognosis is generally good [21]. Many cases, especially in kittens or young
cats, may spontaneously resolve. There is usually a good response to topical 0.1%
tacrolimus or systemic ciclosporin. Some cats may not need further treatment after
resolution, but some may need long-term therapy to maintain remission.
Otitis
207
VetBooks.ir
Para-aural Abscessation
Para-aural abscesses are uncommon in cats. Causes include severely infected
otitis externa and/or media that extends into the surrounding soft tissues, bite
wounds, deep bacterial, mycobacterial and/or fungal infections, complications
from ear canal and/or middle ear surgery, and traumatic ear canal avulsion (usually following a road traffic accident) (Fig. 27). Advanced imaging (especially
contrast-enhanced CT scans) can reveal the extent and severity of the infection
and inflammation, including sinus tracts into adjacent structures and tissues
(which can include the central nervous system). Samples for cytology and culture should be taken from the affected tissues (Fig. 28), as secondary bacterial
infections on the surface may obscure the causative organisms. This may require
surgical exploration.
Treatment depends on the primary cause and may include surgical exploration,
debridement and flushing, vertical ear canal surgery or a total ear canal ablation/
lateral bulla osteotomy. Surgical sites, sinus tracts and debrided tissues should be
thoroughly flushed. Drains may be necessary with deep abscesses or sinus tracts.
Antimicrobials should be selected using culture and administered until clinical
cure. The course of treatment will depend on the extent and severity of infection
Fig. 27 Extensive
para-aural abscesses, ulcers
and sinus tracts in a cat
with an Actinomyces
infection following a bite
wound
VetBooks.ir
208
T. Nuttall
Fig. 28 Deep tissue
cytology from the cat in
Fig. 27. There is
pyogranulomatous
inflammation with a
prominent multinucleated
giant cell. This has several
beaded filamentous
organisms characteristic of
Actinomyces
and inflammation. Prolonged courses aren’t necessary after surgery provided that
the site is clean and closed well, but longer courses (4–6 weeks or longer) will be
necessary with deep infections (especially with slow growing organisms such as
Nocardia, Actinomyces, mycobacteria and fungi).
References
1. Ravens PA, Xu BJ, Vogelnest LJ. Feline atopic dermatitis: a retrospective study of 45 cases
(2001–2012). Vet Dermatol. 2014;25:95.
2. Hobi S, Linek M, Marignac G, Olivry T, Beco L, Nett C, et al. Clinical characteristics and
causes of pruritus in cats: a multicentre study on feline hypersensitivity-associated dermatoses.
Vet Dermatol. 2011;22:406–13.
3. Hensel P, Santoro D, Favrot C, Hill P, Griffin C. Canine atopic dermatitis: detailed guidelines
for diagnosis and allergen identification. BMC Vet Res. 2015;11:196.
4. Miller WH, Griffin CE, Campbell KL. Diseases of eyelids, claws, anal sacs, and ears. In:
Muller and Kirk’s small animal dermatology. 7th ed. St Louis: Elsevier-Mosby; 2013.
p. 723–73.
5. Harvey RG, Paterson S. Otitis externa: an essential guide to diagnosis and treatment. Boca
Raton: CRC Press; 2014.
6. Hammond GJC, Sullivan M, Weinrauch S, King AM. A comparison of the rostrocaudal open
mouth and rostro 10 degrees ventro-caudodorsal oblique radiographic views for imaging fluid
in the feline tympanic bulla. Vet Radiol Ultrasound. 2005;46:205–9.
7. Nuttall TJ, Cole LK. Ear cleaning: the UK and US perspective. Vet Dermatol. 2004;15:127–36.
8. Swinney A, Fazakerley J, McEwan N, Nuttall T. Comparative in vitro antimicrobial efficacy of
commercial ear cleaners. Vet Dermatol. 2008;19:373–9.
9. Buckley LM, McEwan NA, Nuttall T. Tris-EDTA significantly enhances antibiotic efficacy
against multidrug-resistant Pseudomonas aeruginosa in vitro. Vet Dermatol. 2013;24:519.
10. Clark SM, Loeffler A, Schmidt VM, Chang Y-M, Wilson A, Timofte D, et al. Interaction of
chlorhexidine with trisEDTA or miconazole in vitro against canine meticillin-resistant and
-susceptible Staphylococcus pseudintermedius isolates from two UK regions. Vet Dermatol.
2016;27:340–e84.
VetBooks.ir
Otitis
209
11. Pye CC, Yu AA, Weese JS. Evaluation of biofilm production by Pseudomonas aeruginosa from
canine ears and the impact of biofilm on antimicrobial susceptibility in vitro. Vet Dermatol.
2013;24:446–E99.
12. Pye CC, Singh A, Weese JS. Evaluation of the impact of tromethamine edetate disodium
dihydrate on antimicrobial susceptibility of Pseudomonas aeruginosa in biofilm in vitro. Vet
Dermatol. 2014;25:120.
13. Cole LK, Papich MG, Kwochka KW, Hillier A, Smeak DD, Lehman AM. Plasma and ear tissue
concentrations of enrofloxacin and its metabolite ciprofloxacin in dogs with chronic end-stage
otitis externa after intravenous administration of enrofloxacin. Vet Dermatol. 2009;20:51–9.
14. Paterson S. Brainstem auditory evoked responses in 37 dogs with otitis media before and after
topical therapy. J Small Anim Pract. 2018;59:10–5.
15. Greci V, Mortellaro CM. Management of Otic and Nasopharyngeal, and nasal polyps in cats
and dogs. Vet Clin North Am Small Anim Pract. 2016;46:643.
16. Veir JK, Lappin MR, Foley JE, Getzy DM. Feline inflammatory polyps: historical, clinical,
and PCR findings for feline calici virus and feline herpes virus-1 in 28 cases. J Feline Med
Surg. 2002;4:195–9.
17. Greci V, Vernia E, Mortellaro CM. Per-endoscopic trans-tympanic traction for the management of feline aural inflammatory polyps: a case review of 37 cats. J Feline Med Surg.
2014;16:645–50.
18. Janssens SDS, Haagsman AN, Ter Haar G. Middle ear polyps: results of traction avulsion after
a lateral approach to the ear canal in 62 cats (2004–2014). J Feline Med Surg. 2017;19:803–8.
19. Anderson DM, Robinson RK, White RAS. Management of inflammatory polyps in 37 cats.
Vet Rec. 2000;147:684–7.
20. Sula MJM. Tumors and tumorlike lesions of dog and cat ears. Vet Clin North Am Small Anim
Pract. 2012;42:1161.
21. Mauldin EA, Ness TA, Goldschmidt MH. Proliferative and necrotizing otitis externa in four
cats. Vet Dermatol. 2007;18:370–7.
22. Videmont E, Pin D. Proliferative and necrotising otitis in a kitten: first demonstration of T-cell-­
mediated apoptosis. J Small Anim Pract. 2010;51:599–603.
VetBooks.ir
Part III
Feline Skin Diseases by Etiology
VetBooks.ir
Bacterial Diseases
Linda Jean Vogelnest
Abstract
Accurate diagnosis of feline bacterial skin diseases is important for both patient
well-being and appropriate use of antibiotics in times of increasing antimicrobial
resistance. This chapter reviews knowledge of clinical lesions and historical features associated with feline bacterial infections, skin diagnostics relevant to efficient and accurate diagnosis, and current treatment recommendations. Deep
infections including nocardiosis and mycobacteriosis (Chapter, Mycobacterial
Diseases) are well-reported, and although accurate diagnosis is important, and
treatment may be lengthy and challenging, they do occur only rarely. In contrast,
superficial bacterial pyoderma (SBP) is a more common feline presentation that
may be under-recognised, most typically complicating underlying allergic skin
disease, but also associated with a range of underlying diseases and factors. SBP
is reviewed in this chapter, along with deeper infections including deep bacterial
pyoderma, cellulitis and wound abscessation, dermatophilosis, necrotizing fasciitis and environmental saprophytic bacterial infections including nocardiosis.
Confirmation of bacterial skin disease in cats is readily achievable in a general
practice setting. Cytology is often the most valuable tool, used in conjunction
with clues from the history and physical examination and supplemented with
skin surface or tissue culture and/or histopathology when indicated. Cytology
methods relevant to bacterial infections in the cat are detailed in this chapter.
Treatment principles are also discussed, including the potential role of methicillin-­
resistant staphylococci in feline pyoderma, with a focus on current worldwide
recommendations that may supersede some outdated clinic protocols.
L. J. Vogelnest (*)
University of Sydney, Sydney, NSW, Australia
Small Animal Specialist Hospital, North Ryde, NSW, Australia
e-mail: lvogelnest@sashvets.com
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_11
213
214
L. J. Vogelnest
VetBooks.ir
Introduction
Bacterial dermatoses in the cat occur in two broad presentations reflecting the depth
of skin invasion. Superficial infections, involving the epidermis and follicular epithelium, are most common and primarily associated with multiplication of resident
skin microbiota secondary to reduced local and/or systemic host defences. Deep
bacterial infections, involving the dermis and/or subcutaneous tissues, may be extensions of superficial infection or associated with traumatic implantation of a range of
environmental or commensal bacterial species. Some rare but life-­threatening deep
bacterial infections have a propensity for body dissemination.
Normal Feline Cutaneous and Mucosal Bacterial Microbiota
There is limited knowledge about normal commensal bacteria in cats, with most
studies culture-based and focused on staphylococcal isolates. The mouth, followed
by the perineum, appears to be the most consistent staphylococcal carriage site [1].
Fifteen species of staphylococci were identified by MALDI-TOF testing of isolates
from the oropharynx of healthy cats in Brazil, with S. aureus the only coagulase-­
positive staphylococcus (CoPS) species, with a range of coagulase-negative staphylococci (CoNS) [2]. However, α-haemolytic streptococci were more frequently
isolated than staphylococci from healthy mouths of free-roaming cats in Spain, followed by two Proteobacteria (Neisseria spp. and Pasteurella spp.) [3].
Staphylococci have also been less frequently identified as resident skin bacteria
in normal cats, with Micrococcus spp., Acinetobacter spp. and Streptococcus spp.
most common [4]. Of staphylococci isolated, CoNS including S. felis, S. xylosus
and S. simulans have been more frequent than CoPS [4–6], with S. felis potentially
misidentified as S. simulans in some studies [5, 7]. Either S. intermedius (reclassified as S. pseudintermedius in 2005) [1, 8] or S. aureus [5, 9, 10] are variably
reported as the more frequent CoPS isolates. Escherichia coli, Proteus mirabilis,
Pseudomonas spp., Alcaligenes spp. and Bacillus spp. are less frequent isolates
from normal feline skin [4, 5].
More recent genomic DNA studies in healthy cats (n = 11) identified a greater
diversity and number of bacteria on normal feline skin than culture-based studies.
Haired skin had the greatest diversity of species, the pre-aural space the greatest
richness and evenness of species, and mucosal surfaces (nostril, conjunctiva, reproductive) and the ear canal (contrasting to dogs) the lowest species diversity. As for
culture-based studies, Staphylococcus spp. did not dominate, with Proteobacteria
(Pasteurellaceae, Pseudomonadaceae, Moraxellaceae [e.g. Acinetobacter spp.])
most frequent, followed by Bacteroides (Porphyromonadaceae), Firmicutes
(Alicyclobacillaceae, Staphylococcaceae, Streptococcaceae), Actinobacteria
(Corynebacteriaceae, Micrococcus spp.) and Fusobacteria. It is acknowledged that
some species including Propionibacterium spp. may have been under-recognised in
this study [11].
VetBooks.ir
Bacterial Diseases
215
Bacterial residents vary between individuals [4, 11] and may also vary
between healthy and diseased states. Carriage of staphylococci is known to
increase in humans and dogs with atopic dermatitis. Similarly, Staphylococcus
spp. were more frequently detected in allergic cats (n = 10) compared to normal
healthy cats, with more dominance at some anatomic sites (e.g. ear canal) [11].
Staphylococcus spp. were also more prevalent in diseased mouths compared to
normal mouths [3]. In contrast, there was no statistical difference in isolation of
Staphylococcus spp. in another study (n = 98) from healthy skin compared to
inflamed skin [9].
In summary, the feline studies to date suggest, in contrast to dogs, that
Proteobacteria including Acinetobacter spp., Pasteurella spp. and Pseudomonas
spp. are more common on normal feline skin than Staphylococcus spp., and amongst
staphylococci, that CoNS appear to dominate. It is uncertain if staphylococci in general, and CoPS or CoNS in particular, multiply more readily on diseased skin.
Superficial Bacterial Pyoderma
Feline superficial bacterial pyoderma (SBP) is increasingly recognised and reported
in 10–20% of cats presenting to dermatology referral [12–14]. As in other species,
SBP in cats is a secondary disease, most commonly reported with hypersensitivities
[12–14]; 10% of cats presenting to referral in the USA [14] and 60% in Australia
had confirmed underlying allergy, most commonly atopic dermatitis [13]. Recurrent
pyoderma is also commonly reported [13, 15].
Bacterial Species
Although Staphylococcus spp. are considered the likely pathogens [1, 2, 9, 12],
weaker adherence of S. pseudintermedius and S. aureus to normal feline corneocytes in contrast to canine and human corneocytes has been documented [16], and
the casual bacterial species in feline SBP have only been confirmed in a small
number of cats. S. aureus was isolated in pure culture from papules and crusts of
one cat, with concurrent neutrophils on skin cytology, and complete resolution of
lesions by 10 days of antibiotic therapy [17]. S. felis was isolated from the nostrils
and skin lesions (excoriations) of another cat with suspected underlying flea bite
hypersensitivity, with concurrent neutrophils and intracellular cocci on cytology,
and complete resolution of lesions by 14 days of antibiotic therapy and flea control [5]. Eosinophilic granuloma complex lesions may also be complicated by secondary pyoderma, and the most common isolates from surface swabs and/or tissue
biopsies from eosinophilic plaques or lip ulcers (n = 9), with concurrent neutrophils
and intracellular cocci on cytology, were S. pseudintermedius and S. aureus. Other
isolates detected in this study included CoNS, Pasteurella multocida, Streptococcus
canis and Pseudomonas aeruginosa [12].
VetBooks.ir
216
L. J. Vogelnest
A number of other bacterial culture studies, predominantly on laboratory isolates from a range of skin lesions unconfirmed as pyoderma, have focused on
staphylococci; whether isolates were pathogenic or incidental is uncertain, and non-­
staphylococcal isolates are rarely reported [4, 7, 9, 17–19]. CoNS are the most common isolates in a number of studies, accounting for 96% of isolates from ‘inflamed
skin’ (n = 24) [9], the second most frequent isolate (S. simulans) from abscesses,
miliary dermatitis, excoriations, exfoliative dermatitis or eosinophilic plaques
(n = 45) [17] and the most frequent isolates (S. felis followed by S. epidermidis)
from unspecified ‘dermatitis’ [7]. Less common CoNS isolates include S. hyicus, S.
xylosus and S. schleiferi subsp. schleiferi [9, 17].
CoPS have been more prevalent in some studies on diseased feline skin [4], with
S. aureus (n = 69) [9, 17] or S. intermedius (n = 9 [5]; n = 30 [20]) the most frequent
isolates, and Streptococcus spp. (10%), Proteus spp. (10%), Pasteurella spp. and
Bacillus spp. (10%) also reported [20].
The relative importance of staphylococci in general, and CoNS and CoPS in
particular, to feline pyoderma and whether there is one predominant causal species
as for bacterial pyoderma in humans (S. aureus) and dogs (S. pseudintermedius) is
currently uncertain.
Clinical Presentation
A median age of onset of 2 years is documented for feline SBP, although a wide
range is reported (6 months to 16.5 years), with older cats also frequently affected
(first presentation at >9 years of age in 23% of cats) [13]. Pruritus is common,
particularly with underlying hypersensitivity, reported in 92% of cats with SBP in
Australia and often severe (56%) [13]. Lesions associated with feline SBP often
reflect self-trauma, consisting most typically of multifocal, crusted, alopecic,
excoriated and erosive to ulcerative lesions (Figs. 1, 2, 3 and 4). Eroded papules,
eosinophilic plaques, eosinophilic granulomas and rare pustules are also reported.
The most frequent lesional sites are the face, neck, limbs and ventral abdomen
[12, 13, 21].
Diagnosis
Although some clinical lesions have been recognised as useful diagnostic clues
for bacterial pyoderma in dogs [22, 23], SBP lesions in cats are less characteristic, with many non-specific presentations (e.g. erosions, crusting). Diagnostic tests
are thus important to confirm a diagnosis of feline pyoderma (see later section on
“Cytology”, Table 1) and are strongly encouraged prior to consideration of treatment with systemic antimicrobials [22–24].
Cytology has been considered the most useful single test, with the presence
of neutrophils and intracellular or associated bacteria being diagnostic (Fig. 11a)
VetBooks.ir
Bacterial Diseases
217
Fig. 1 Feline secondary
bacterial pyoderma (SBP):
exudative erosions and
crusting
Fig. 2 Feline SBP:
alopecia, erythema and
focal crusting
[12, 13, 22, 25]. In canine pyoderma, cytology is considered mandatory when
typical lesions (pustules) are not present or scant and is also essential to identify
concurrent or alternate Malassezia dermatitis [23]. The morphology of bacteria on
cytology (cocci and/or rods) will also guide valid empirical treatment choices and/
or the need for bacterial culture. Adhesive tape impressions are applicable to all
superficial skin lesions, in particular dry lesions and restricted body sites, while
glass slide impressions are suitable for erosive to ulcerative lesions [22]. In canine
SBP, it is reported that inflammatory cells and bacteria may be absent or scarce with
concurrent immunosuppression from disease or drugs [23].
Histopathology is infrequently discussed in relation to diagnosis of SBP;
however, it can provide further diagnostic confirmation, especially if samples are
VetBooks.ir
218
L. J. Vogelnest
Fig. 3 Feline SBP:
erythematous eroded
plaques
Fig. 4 Feline SBP:
well-demarcated alopecia
and erythema with focal
crusting
collected without prior skin surface cleansing or disinfection as bacterial colonies are
frequently observed within the crusts (Fig. 5) (see later section on Histopathology).
Histopathology is also valuable to aid exclusion of other differentials for atypical
presentations or when a diagnosis is uncertain [22].
Bacterial culture is not helpful for diagnosis of SBP, particularly when assessed
independently of cytology, as isolation may simply reflect normal commensal species not involved in disease (see later section on Bacterial Culture) [6]. A heavy
pure culture of one bacterial species is more likely associated with a pathogen than
mixed-species isolation, but concurrent cytology remains essential [1]. Coagulase
status of any staphylococci isolated is less helpful for feline pyoderma, as both
CoNS and CoPS are potentially pathogenic. Despite a limited role diagnostically,
culture and antibiotic susceptibility testing (C&S) can be important to guide appropriate antibiotic therapy, particularly when antimicrobial resistance is more likely.
Bacterial Diseases
219
VetBooks.ir
Table 1 Differential diagnoses and valuable diagnostic tools for cutaneous lesions associated
with bacterial infections in cats
Less common
differentials for lesion
Ectoparasites (Otodectes,
larval ticks,
trombiculids);
pemphigus foliaceus
Lesion
Papules
Common differentials
SBP, allergya
dermatophytosis
Alopecia,
erythema,
scaling,
crusting
SBP, dermatophytosis,
allergya, actinic
keratoses (non-­
pigmented skin)
Demodicosis (D. gatoi,
D. cati), pemphigus
foliaceus, ectoparasites
(Cheyletiella, lice)
Erosion,
ulceration,
crusting
SBP, allergya, SCC
(non-pigmented skin)
Herpes viral dermatitis,
SCC in situ, cutaneous
vasculitis
Erythematous
plaques
SBP, allergya
Cutaneous xanthoma
Nodules (lip,
chin, linear)
Nodules
(poorly
demarcated)
Nodules
(discrete)
SBP, DBP, allergya
Mycetoma, neoplasia
(SCC)
Mycobacteria, Nocardia,
sterile panniculitis
Pustules (rare)
SBP, pemphigus
foliaceus
Bacterial cellulitis/
abscessation
Neoplasia (variety),
eosinophilic
granuloma
Pseudomycetoma
(bacterial,
dermatophyte),
mycetoma, histiocytosis,
sterile pyogranuloma
Dermatophytosis
Diagnostic tools
History (parasiticides,
exposure/contagion),
cytology (tape
impression), biopsy
(histo)
History (potential
exposure/contagion,
pruritus or lesions
first), cytology (tape
impression), biopsy
(histo)
History (degree of
pruritus, recurrent/
seasonal), cytology
(tape or slide
impression), biopsy
(histo)
Cytology (tape or slide
impression), biopsy
(histo)
Cytology (FNA),
biopsy (histo)
Cytology (FNA),
biopsy (histo, C&S)
Cytology (FNA),
biopsy (histo, C&S)
Cytology (impression
after rupture), biopsy
(histo)
Atopic dermatitis, adverse food reactions and/or flea bite hypersensitivity
C&S culture and antibiotic susceptibility testing, DBP deep bacterial pyoderma, FNA fine needle
aspirates, histo histopathology, SBP superficial bacterial pyoderma, SCC squamous cell carcinoma
a
Treatment
There are limited studies evaluating treatment of feline SBP, and most recommendations are anecdotal. However, recent guidelines stress the importance of confirming a
diagnosis of SBP prior to considering systemic antibacterial therapy (see later section on
Antibiotic Stewardship (Box 1)) [1, 22, 23]. Over-utilisation of antibiotics without confirmation of diagnosis is well-recognised, and the common practice of prescribing antibiotics ‘just in case’ is strongly discouraged [22–24, 26]. Topical antiseptic therapy is
a more valid ‘just in case’ choice; however, prior cytology is always recommended [1].
VetBooks.ir
220
L. J. Vogelnest
Fig. 5 Bacterial colonies,
usually cocci, are
frequently observed in
biopsies from feline
cutaneous lesions with
secondary bacterial
infection (H&E, 400×).
(Courtesy of Dr. Chiara
Noli)
Topical Therapy
Although cats are often considered less tolerant of topical therapies, and even in
dogs topical therapy is considered under-utilised [23], topical therapy has been
recommended as the optimal sole antibacterial treatment for superficial infections whenever achievable for the pet and owner, particularly for localised or
mild lesions. It is also recommended as the best option for pyoderma associated with methicillin-­resistant staphylococci (MRS) [1]. Topical therapy has the
advantage of more rapid lesion resolution, reduced duration of systemic antibiotics, physical removal of bacteria and debris from the skin surface and reduced
impact on bystander commensals [1, 23]. The response in dogs with SBP to daily
chlorhexidine spray (4%) for 4 weeks concurrently with twice weekly bathing
with chlorhexidine shampoo was comparable to oral amoxicillin-clavulanic acid
(amoxi-clav) [27]. Other small studies have similarly shown sole topical therapy
to be effective [1].
Although a range of topical formulations are discussed for use in dogs, it is
acknowledged there is limited evidence for efficacy and safety to guide optimal
choices and protocols [23]. There is even less evidence in cats. However, the author
has found a range of topical antiseptics and antibiotics helpful in the treatment of
SBP in some cats, particularly for localised lesions. Chlorhexidine solution (2–3%
VetBooks.ir
Bacterial Diseases
221
Box 1: Important Principles of Treatment for Cutaneous Bacterial Infections in
Cats in Line with Good Antimicrobial Stewardship
• Have sufficient evidence to confirm a diagnosis of bacterial infection prior
to instigating treatment (unless severe and life-threatening): Avoid ‘just-in-­
case’ usage.
–– Cytology is essential; culture of bacteria from a skin surface swab does
not confirm infection.
• Choose antibiotics wisely, based on recommended treatment guidelines:
–– Use first-line antibiotics for empirical use, assuming relevant options
exist for the confirmed infection.
–– Only use second-line antibiotics if adverse events limit use of first-line
choices and if culture and sensitivity testing (C&S) supports efficacy.
–– Do not use third-line antibiotics (e.g. cefovecin, fluoroquinolones)
unless C&S indicates absence of other first- or second-line choices:
Avoid justification due to ‘ease of use’ without actively discussing first-­
line oral alternatives.
• Use correct dose and duration of treatment:
–– Dose at the upper end of dose range as skin blood supply is comparatively poor, and weigh patients: slightly over-dose rather than
under-dose.
–– Follow duration guidelines for the confirmed infection, and re-evaluate
clinical and cytological response prior to cessation of therapy.
once or twice daily), silver sulfadiazine 1% cream or mupirocin 2% ointment (twice
daily) have apparent efficacy and safety [12, 13], and fusidic acid 1% viscous eye
drops (Conoptal®; twice daily) may also be useful, particularly for facial/periocular
lesions. Concern has been raised over the use of both mupirocin and fusidic acid
in veterinary patients, potentially encouraging resistance in resident human staphylococci, and it has been recommended to restrict their use to cases without other
practical choices [1, 23]. Shampoo therapy (chlorhexidine or piroctone olamine)
once to twice weekly may be adjunctive for treatment or to inhibit recurrence of
SBP, although it is poorly tolerated in many cats.
Excessive grooming and exacerbated self-trauma in response to topical therapies
in cats, especially to ointments or creams, may sometimes limit their use. Body suits
or conforming bandages may be helpful, particularly in cats with severe pruritus.
Despite a common concern of owners that licking will remove topical medications,
there is no evidence to confirm that grooming notably reduces efficacy of topical
therapy, as lipophilic medications will be quickly absorbed after application.
Systemic Therapy
There is a lack of consensus on the most appropriate systemic antibiotics for treatment of SPB and some variation in recommendations with geographical region
[23, 28]. First-line antibiotics are considered suitable choices for empirical therapy,
VetBooks.ir
222
L. J. Vogelnest
assuming a diagnosis is confirmed (e.g. intracellular cocci on cytology). Culture and
antibiotic susceptibility testing (C&S) is important for cases that respond poorly to
appropriate empirical therapy, or if there is higher risk of MRS (repeated antibiotic
courses, other household pet carriers, some geographical regions) [1, 12].
Amoxi-clav and cephalexin are generally considered first-line choices for feline
SBP (see later section on Antibiotic Stewardship) [12, 13]. Amoxi-clav was effective for eosinophilic plaques and partially effective for lip ulcers with concurrent
bacterial infection [25]. Doxycycline is used in some countries for first-line therapy
of SBP, but resistance in some geographical regions [29], and potential value for
MRS and multidrug-resistant staphylococci in others [10], suggests it may be less
appropriate for first-line use. There is also debate over the use of cefovecin as first-­
line treatment for feline SBP, and although it is commonly adopted, third-generation
cephalosporins are considered critically important antibiotics in human medicine,
reserved for life-threatening diseases [26, 30–32]. It has thus been recommended
cefovecin is not appropriate for first-line treatment for feline SBP, unless, due to
compliance issues, no other treatment is possible.
Second-line antibiotics may be considered if first-line antibiotics are not effective or serious side effects (real or potential due to previous history) limit the use
of first-line choices. The major second-line choices for feline SBP are clindamycin
or doxycycline, with preceding C&S optimal as efficacy is less predictable than
for first-line choices (see later section on Antibiotic Stewardship). Lower sensitivity of staphylococcal isolates has been documented to clindamycin compared to
amoxi-­clav and cephalexin in South Africa [8] and to erythromycin in Malaysia
[29]. Cefovecin is another potential second-line choice, when all avenues of oral
administration of first-line and initial second-line choices have been exhausted.
Second-­generation fluoroquinolones (FQ) (enrofloxacin, marbofloxacin) are a final
consideration, but restriction to cases with no other alternatives based on C&S is
recommended. Ease of administration of FQ and low incidence of side effects are
not justification for their use as first-line or early second-line options.
Third-line antibiotics are rarely indicated for feline SBP, with topical therapies,
even requiring hospitalisation and/or sedation where necessary, preferable. They
include third-generation FQ (orbifloxacin, pradofloxacin), aminoglycosides (amikacin, gentamicin) and rifampicin. Critical antibiotics, reserved for life-threatening
infections in humans, with veterinary use discouraged, are not a consideration for
treatment of SBP in any species (see later section on Antibiotic Stewardship).
Duration of Therapy
Although there is an absence of scientific evidence to confirm an optimal duration
of therapy for SBP in either dogs or cats, current expert opinion recommends a
3-week therapy as most appropriate [1, 26]. Shorter courses may be considered,
until clinical lesions and microbiological evidence of infection have resolved; however, re-evaluation of patients is essential to make this assessment [1, 28].
VetBooks.ir
Bacterial Diseases
223
reatment of the Primary Disease
T
It is well-recognised that the underlying primary cause of SBP must be managed to
limit recurrence. However, there is less clarity on whether treatment of SBP and primary diseases need to occur concurrently or sequentially. As immunosuppressive therapy is contraindicated when treating infectious diseases, as a general rule, it is advised
that SBP treatment be completed, prior to commencing any sustained glucocorticoid
therapy (e.g. for primary allergy). In some cases of very active primary disease, resolution of SBP may not readily occur until the primary disease is more controlled.
Management of primary atopic dermatitis in particular can be very challenging in some
cats prone to secondary bacterial infections [13]. Ciclosporin therapy may be a more
valid allergy treatment choice than glucocorticoids in this scenario, sparing innate
immune responses (neutrophils, macrophages), albeit with slower onset of effect.
Deep Bacterial Infections
Chin Nodular Swelling: Secondary Deep Bacterial Pyoderma
Feline chin acne most typically presents with brown to black comedones and hair
casts on the ventral chin and occasionally the margins of the lower or upper lips
(Chapter, Idiopathic Miscellaneous Diseases). A proportion of affected cats develop
notable swelling with draining tracts, often due to secondary deep bacterial infection. Of cats with feline acne presenting to referral hospitals in the USA, 42% had
deep bacterial infection (n = 72) [33], and 45% had bacteria isolated from tissue
cultures (n = 22), including all cats with evidence of folliculitis and furunculosis on
histopathology. The most frequent bacteria isolated, typically in pure culture, were
CoPS, followed by α-haemolytic streptococci, Micrococcus sp., E. coli and Bacillus
cereus. Of note, Pseudomonas aeruginosa was isolated in heavy growth from the
tissue biopsy of one healthy control cat [34].
Clinical Presentation
Deep pyoderma typically presents with large papules to nodular swelling with
draining tracts (Fig. 6) and less commonly diffuse swelling. Lesions may be pruritic
and/or painful, and regional lymph node enlargement can occur [3, 33, 34].
Diagnosis
Cytology from fine needle aspirates (FNA) or expressed discharge after initial surface cleansing may reveal intracellular bacteria within neutrophils and/or macrophages. Careful examination may be required as bacteria can be sparse in samples
from nodular lesions despite marked inflammation.
Histopathology will typically reveal folliculitis, furunculosis and perifollicular
to nodular pyogranulomatous inflammation (Fig. 7); bacteria present within follicular ostia or lumina in this setting, at least focally, confirm a diagnosis. Feline acne
VetBooks.ir
224
L. J. Vogelnest
Fig. 6 Feline chin acne:
nodular swellings and
drain tracts as a
consequence of a deep
bacterial infection.
(Courtesy of Dr. Chiara
Noli)
Fig. 7 Histopathological
section from feline chin
acne (H&E, 40×):
multifocal nodular
pyogranulomatous
inflammation in the mid
and deep dermis, mostly
centred on the hair
follicles, which appear
completely destroyed.
Haemorrhage is evident,
which is reflected clinically
by haemopurulent exudate.
(Courtesy of Dr. Chiara
Noli)
is associated with a spectrum of histopathology changes, with periglandular and/
or perifollicular inflammation usually dominating. Sebaceous gland ductal dilation
and pyogranulomatous inflammation of sebaceous glands are also reported [34].
VetBooks.ir
Bacterial Diseases
225
The presence of folliculitis and furunculosis without causal bacteria is suggestive
of a role for secondary bacterial pyoderma, but exclusion of other causes including
dermatophytosis is important, and special stains are warranted.
Bacterial culture of sterile tissue biopsies or FNA from affected regions is
required to identify causal species and enable antibiotic susceptibility testing.
Treatment
Systemic antibiotics are indicated; if intracellular cocci are evident on cytology,
empirical treatment with cephalexin or amoxi-clav is often considered suitable. If
bacterial rods are present on cytology, or in geographical regions where MRSP is
more common, C&S is recommended, optimally from tissue biopsies. The optimal
duration of therapy for deep pyoderma is undetermined; however, a minimum of
4–6 weeks is often advised, continuing for at least 2 weeks beyond resolution or stasis of lesions [1, 26]. Comedones typically persist in feline acne following resolution
of the bacterial infection, so further treatment of the underlying pathology is important to limit recurrent infection (Chapter, Idiopathic Miscellaneous Diseases) [33].
Discrete Nodules: Bacterial Pseudomycetoma
Some bacteria rarely cause localised discrete deep infections forming skin nodules that mimic fungal or neoplastic causes. Infections presumably occur following
traumatic implantation of bacteria, which are most commonly Staphylococcus spp.,
but may be Streptococcus spp., Pseudomonas spp., Proteus spp. or Actinobacillus
species.
Clinical Presentation
Single or multiple inflammatory nodules, with or without draining tracts, are typical.
Discharge may contain small white grains or granules, composed of compact bacterial colonies [35]. A single case with less typical overlying thick crusting is reported
in an FIV-positive cat, with concurrent SBP supported by cytology findings [36].
Diagnosis
Cytology of FNA from intact nodules or impression smears of freshly expressed
exudate should reveal numerous bacteria, most typically cocci but dependent
on causal species. Histopathology will reveal nodular to diffuse pyogranulomatous dermatitis and/or panniculitis with numerous macrophages, multinucleate
giant cells and central aggregations of bacteria, often with a brightly eosinophilic amorphous periphery (Splendore-Hoeppli phenomenon) (Figs. 8 and 9)
[35, 36].
Treatment
Surgical excision/drainage is important for resolution, as systemic antibiotics will
often not penetrate into the central walled-off bacteria.
VetBooks.ir
226
L. J. Vogelnest
Fig. 8 Histopathological
section from a lesion of
bacterial pseudomycetoma
(H&E 40×). There is a
multifocal nodular
pyogranulomatous
inflammation with large
bacterial colonies covered
by bright red proteinaceous
material, which appear
clinically as white granules
in the exudate. (Courtesy
of Dr. Chiara Noli)
Fig. 9 A bacterial colony
(dark blue in the centre, is
surrounded by amorphous
eosinophilic material
(Splendore-Hoeppli
phenomenon) (H&E,
400×). (Courtesy of Dr.
Chiara Noli)
ubcutaneous Nodular Swellings with Abscessation: Anaerobic
S
Bacteria
Painful rapidly progressing subcutaneous swellings are common in cats due to
implantation of anaerobic bacteria, most typically associated with fight wounds
although less commonly with other skin trauma including surgical wounds or catheterisation. Causal bacteria are often anaerobic or facultatively anaerobic oral commensals, including Pasteurella multocida, Fusobacterium spp., Peptostreptococcus
spp., Porphyromonas spp. and gas-producing species such as Clostridium spp. and
Bacteroides species [37].
VetBooks.ir
Bacterial Diseases
227
Fig. 10 Swelling,
ulceration, fistulisation and
necrosis of the abdominal
skin of a cat due to
infection with anaerobic
bacteria. (Courtesy of Dr.
Chiara Noli)
Clinical Presentation
Poorly demarcated areas of oedema and swelling are typical, which progress to
abscessation (Fig. 10) and sometimes overlying skin necrosis. Lesions are often single, but may be multiple, and are usually painful. There is often associated pyrexia
and malaise, especially with larger lesions or when bacteria produce toxins. Purulent
abscess contents often have a putrid smell, and tissue crepitus may be apparent.
Diagnosis
The clinical presentation is usually diagnostic. Cytology of abscess contents, or
FNA from oedematous areas in early lesions, should reveal intense neutrophilic
inflammation, with bacterial rods and/or cocci often readily apparent. Mixed infections are not unusual. Culture is generally not required, but anaerobic sampling
would be important to accurately identify most causal bacteria.
Treatment
Early lesions are usually managed successfully with systemic antibiotics, with most
organisms sensitive to amoxi-clav or metronidazole. Bacteroides spp. may be resistant to ampicillin and clindamycin [30]. Surgical drainage of abscesses, with aeration and cleansing of infected tissue, is important to resolution.
ubcutaneous Nodular Swellings with Ulceration and Draining
S
Tracts: Nocardia, Rhodococcus and Streptomyces
A number of bacterial species, many of which are ubiquitous environmental saprophytes, are rare causes of poorly demarcated nodular swellings with focal ulceration
and draining tracts in cats. Infections are often locally invasive, and some species
have a propensity to disseminate, particularly in immunocompromised cats. Most
infections presumably occur following traumatic implantation.
VetBooks.ir
228
L. J. Vogelnest
Diagnostic tests are essential to accurately confirm the cause of this presentation.
In addition to multiple potential bacteria, differential diagnoses include mycobacteria
(Chapter, Mycobacterial Diseases), saprophytic fungi (Chapter, Deep Fungal Diseases)
and sterile panniculitis (Chapter, Idiopathic Miscellaneous Diseases).
Cytology of FNA from oedematous tissue or fluid pockets or of smears from
draining tracts (after initial skin surface cleansing) will typically reveal neutrophils
and epithelioid macrophages, sometimes with multinucleate giant cells, regardless
of the causal organism. Organisms will more often be detected within macrophages,
with morphology varying with the causal species.
Histopathology of tissue biopsies will reveal nodular to diffuse pyogranulomatous dermatitis and/or panniculitis. Specials stains help elucidate the likely causal
bacteria [38].
Bacterial culture from sterile fluid aspirates or tissue biopsies may be needed
to confirm causal species and is optimal to determine antimicrobial susceptibility
testing. It is important to alert the laboratory of the potential for unusual bacterial
species with special culture requirements.
PCR testing can be useful for retrospectively identifying pathogens from formalin-­
fixed tissue samples if fresh samples are not available for bacterial culture [39].
Nocardiosis
Nocardia are ubiquitous soil and decaying vegetation saprophytes that may cause
rare but potentially serious infection in cats, typically following implantation into
skin wounds. Infection is more common in cats than dogs and may remain localised
and indolent or be fulminant with wide dissemination; the latter course is more
likely in immunocompromised hosts. N. nova is the most frequently identified
causal species, but infections with N. farcinica or N. cyriacigeorgica also occur.
Skin infections are most common, with occasional cases restricted to pulmonary or
abdominal infection [40].
Clinical Presentation Progressive irregular nodules and punctate draining sinuses
are typical (Fig. 11), often with concurrent malaise and respiratory signs. Skin
infection may start with discrete abscesses that gradually extend into discharging,
non-healing wounds. The extremities, ventral abdomen and inguinal areas are more
often affected, and lymphadenopathy is common. Discharge may contain gritty
granules (bacterial microcolonies) [40].
Diagnosis Filamentous bacteria that stain at least partially with acid-fast stains are
typically prevalent on cytology and histopathology and appear branching or beaded
(Fig. 12). Organisms may be found within clear lipid vacuoles [40]. Bacterial culture is slow; it is important to forewarn laboratories with potential cases.
Treatment Prompt early treatment of acute lesions, even in immunocompromised
patients, can result in good outcomes. Surgical debridement and drainage to reduce residual organisms are optimal, and aggressive early excision, with potential later corrective
surgery, is indicated. C&S is important to maximise treatment success. N. nova tends to
have less resistance than other species and is often susceptible to sulphonamides,
VetBooks.ir
Bacterial Diseases
229
Fig. 11 Localised
swelling, ulceration and
training tract in a cat
affected by cutaneous
nocardiosis. (Courtesy of
Dr. Carolyn O’Brien)
Fig. 12 Cytology of
nocardiosis: multiple
groups of bacteria (grains)
and slender and
filamentous Nocardia
asteroides microorganisms
are evident (MGG 1000×).
(Courtesy of Dr. Nicola
Colombo)
tetracyclines (minocycline, doxycycline), clarithromycin and ampicillin/amoxicillin, but paradoxically not to amoxi-clav (clavulanic acid induces β-lactamase production in these species) nor to FQ. Amoxicillin (20 mg/kg twice daily) combined
with clarithromycin (62.5–125 mg/cat twice daily) and/or doxycycline (5–10 mg/kg
twice daily) is recommended over sulphonamides. Long-term therapy is generally
required (3–6 months), and recurrence is common with shorter treatment. N. farcinica is less commonly identified but is often multidrug resistant and highly pathogenic. Initial parenteral therapy with amikacin and/or imipenem combined with
trimethoprim-sulphonamides is a consideration [40].
Rhodococcosis
Rhodococcus equi is a ubiquitous soil-borne bacterium commonly pathogenic in
horses, where it produces a pyogranulomatous pneumonia and enteritis with high mortality in young foals. Infection is also increasingly documented in humans with immunocompromise and is reported in a small number of cats, involving skin (nodules with
VetBooks.ir
230
L. J. Vogelnest
focal ulceration and draining tracts, most frequently on the extremities), abdominal or
thoracic cavities and/or the respiratory tract [41–43]. In one report, a pyogranulomatous skin disease and cellulitis (Fig. 13), different from usual presentations in cats, were
described in a 2-year-old female domestic shorthaired cat [43]. Infection in local lymph
nodes, presumably via lymphatic spread, is reported [41–43]. Implantation of organisms via skin wounds is proposed, with highest risk in cats with exposure to horses;
infected foals shed copious bacteria into the environment via faeces [41].
Diagnosis Cytology of FNA samples and/or histopathology usually readily reveals
gram-positive cocci to coccobacilli within macrophages (Fig. 14) [42, 43]. Bacterial
culture is essential to confirm a diagnosis; the bacteria grow readily with aerobic
culture within 48 hours, but organisms may be protected within macrophages in
fluid samples, so tissue samples may be optimal [42].
Treatment C&S is important to guide potential therapy. R. equi infections are
often refractory to conventional therapies in horses, and although a combination of
rifampicin and erythromycin has been recommended, increasing resistance is recFig. 13 Cutaneous
Rhodococcus equi infection
in a cat: pyogranulomatous
dermatitis and cellulitis with
superficial ulceration.
(Courtesy of Dr. Anita Patel)
Fig. 14 Cytology of case
in Figure 13: intracellular
Rhodococcus equi
organisms are evident in
macrophages (MGG
1000×). (Courtesy of Dr.
Anita Patel)
VetBooks.ir
Bacterial Diseases
231
ognised [28]. In a confirmed feline case with a chronic limb lesion, R. equi displayed intermediate sensitivity to amoxi-clav, rifampicin and erythromycin and
sensitivity to cephalexin and gentamicin, but the cat deteriorated despite initial
cephalexin and later surgical debridement and gentamicin therapy and was
euthanised [42]. In another case with sensitivity reported to doxycycline, enrofloxacin and cefuroxime, response to enrofloxacin and later doxycycline was poor [43].
However, doxycycline was reported effective in thee kittens with R. equi pneumonia, from two litters in a cattery in Australia where the source of the infection was
undetermined [41].
Streptomycosis
Streptomyces spp. are ubiquitous environmental bacteria that very rarely cause
irregular nodular lesions with draining tracts and dark tissue granules on the limbs
and ventral abdomen of cats. One cat without skin lesions had mesenteric and lymph
node infection. Two cats were FIV and/or FeLV positive and two cats had unknown
viral status [38].
Diagnosis Gram-positive rods to coccobacilli were present on cytology and histopathology, and bacteria were identified by PCR testing [38].
Treatment All four cats failed to respond to surgical and/or multiple antibiotic
therapy and were euthanised following 6–18 months of disease [38].
Dermatophilosis
Dermatophilosis is a contagious and potentially zoonotic disease caused by
Dermatophilus congolensis, which most commonly affects cattle, sheep and horses
in tropical and subtropical climates. The organism does not survive readily in the
environment, and infected or carrier animals are the main source. Infection is
reported very rarely in cats. Two presumptive cases presented with nodular swelling
and draining tracts overlying infected lymph nodes, with associated skin surface
crusting. Characteristic gram-positive branching filamentous bacteria were evident
on histopathology, and both cats resided on farms in tropical northern Australia.
Surgical excision was curative in one cat, and the other cat was euthanised prior
to diagnostics [44]. Dermatophilus congolensis was isolated in pure culture from
crusts in another cat presenting with crusting and exudation on the ventrolateral lip
margins; it was reported sensitive to oxytetracycline and penicillin, but resistant
to ampicillin, amoxicillin, gentamicin and cefoperazone [45]. Characteristic filamentous branching bacteria (Fig. 15) were present on cytology from a fourth cat
with draining tracts on two lower limbs; bacterial culture was negative, but the cat
responded completely to amoxicillin therapy for 10 days [46].
Streptococcal Infection
One case of extensive oedematous swelling with multifocal ulceration and draining
tracts is reported on the hindlimb of a cat, associated with numerous clusters and
chains of gram-positive cocci, identified by tissue PCR as Streptococcus spp., in skin
VetBooks.ir
232
L. J. Vogelnest
Fig. 15 Cytology of
Dermatophilus
congolensis: long colonies,
like train tracks, are
characteristic
(Diff Quik, 1000x)
and underlying bone. Clusters of bacteria surrounded by eosinophilic amorphous
material (Splendore-Hoeppli phenomenon) were present on histopathology [39].
Actinomycosis
Actinomyces spp. are oral saprophytes in a variety of animals including dogs and
cats, which are most commonly associated with soft tissue and bone infections in
the jaws of cattle. Rare cutaneous infections are reported in dogs, characterised by
nodular swellings with discharge, typically on the extremities. Although abdominal infection with Actinomyces spp. is documented in one cat, and isolation of
Actinomyces spp. is reported concurrently with other bacterial species, or from
lesions without concurrent histopathological confirmation, there are no confirmed
reports of Actinomyces spp. causing cutaneous infections in cats [47, 48].
apidly Progressive Oedematous Swelling to Necrosis and Septic
R
Shock: Necrotizing Fasciitis
Necrotizing fasciitis is a rapidly progressive and frequently fatal syndrome caused
by severe bacterial infection of subcutaneous tissue (fascia) and adjacent skin,
typically associated with septic shock. Streptococcus canis is a recognised cause
of fulminant disease in humans and dogs and has also been associated with an
outbreak of fatal necrotizing fasciitis in shelter cats in southern California. Clonal
bacteria were identified and spread via close physical contact was proposed. S.
canis is a normal inhabitant of the urinary, reproductive and gastrointestinal tracts
of dogs and cats, and although infections are rare and most typically associated
with immunocompromise, necrotizing fasciitis can occur in immunocompetent
hosts. In contrast to dogs where S. canis is mainly associated with skin infections,
respiratory tract infections are more typical in cats [49]. One case associated with
S. canis in a single cat following minor limb trauma is also reported [50].
VetBooks.ir
Bacterial Diseases
233
Fig. 16 Large areas of
necrosis and ulceration in a
cat with necrotising
fasciitis. (Courtesy of Dr.
Susan McMillan)
Another form of necrotizing fasciitis in people, occurring after minor skin
trauma (catheterisation, hospitalisation), has been associated with multiple concurrent bacteria including Staphylococcus spp., Streptococcus spp., Pseudomonas spp.
and E coli. Single case reports in cats are described due to Acinetobacter baumannii
[51] and multiple bacteria (E. coli, Enterococcus sp. and S. haemolyticus; E. coli,
Enterococcus faecium and S. epidermidis) [52, 53].
Clinical Presentation Poorly demarcated painful regions of oedema and erythema
are typical, associated with rapid development of signs of septic shock (pyrexia,
severe malaise, collapse). Skin lesions progress to large areas of skin necrosis (Fig. 16).
Diagnosis FNA of affected regions reveals neutrophilic inflammation, and causal bacteria are usually apparent intracellularly within neutrophils. Bacterial culture of sterilely
collected fluid or tissue samples is required to confirm the causal species. It is important
to interpret culture results in conjunction with bacterial morphology from cytology and/
or histopathology, as contaminant species may be cultured from exudative lesions.
VetBooks.ir
234
L. J. Vogelnest
Treatment Most cases reported in cats have been fatal. Urgent extensive surgical
debridement, with removal of the bacterial nidus and all necrotic tissue to limit
further extension along fascial planes, is recognised as crucial for suspected cases
prior to availability of diagnostic test results, together with broad-spectrum intravenous antimicrobial treatment and critical care. Reconstructive surgery may be
required after recovery [50].
Diagnostic Tools for Feline Cutaneous Bacterial Infections
Clinical Lesions and Historical Features
Prior to reaching for diagnostic tests, careful clinical examination and history taking
for each case can focus the diagnostic possibilities and guide the most appropriate
test choices. Knowledge of the more likely differentials for specific skin lesions,
and the major differentials when bacterial infections are being considered, is helpful
(see Table 1).
Cytology
Cytology is often the most useful initial test when considering bacterial dermatoses
and may confirm a diagnosis. The most suitable technique will vary with the clinical
lesions (see Table 1).
Adhesive tape impressions are suitable for all superficial skin lesions,
including alopecia, scaling, crusting, excoriations, ulceration and papules. More
exudative lesions can be gently blotted with a dry gauze swab prior to sampling.
Good quality adhesive tape (clear, transparent, strongly adhesive; 18–20 mm
width) is optimal for use on standard glass slides. Tape strips (~5–6 cm long) are
pushed firmly onto lesional skin, squeezing gently on intact papules or plaques
and repositioning repeatedly until adhesiveness reduces. Tapes are stained with
a Romanowsky stain (e.g. Diff-Quik®) without initial fixation (dissolves the
adhesive, reducing clarity). Use of the red stain is useful in cats to aid identification of eosinophils. Tapes can be dipped into stain pots, as for glass slides
(Fig. 17).
Glass slide impressions are suitable for moist lesions, including erosions and
ulcers, and for sampling pustules after rupture with a sterile needle. Slides are
stained with a Romanowsky stain including the fixative. Heat fixing is not required.
Fine needle aspirates are suitable for deeper lesions, including larger papules
and nodules. The skin surface should be gently disinfected with alcohol prior to
sampling. Aspirated samples are quickly sprayed from the hub of the needle onto
glass slides using an air-filled syringe. Slides are air-dried prior to routine staining
with a Romanowsky stain or with gram and/or acid-fast stains for identification of
less common bacterial species.
Bacterial Diseases
b
c
VetBooks.ir
a
235
d
e
f
Fig. 17 Staining of an adhesive tape impression: (a) after sample collection, the tape is pressed
firmly at one end, adhesive side down, onto a glass slide and curled into a slightly offset cylinder;
(b) tape is dipped into red stain of Diff-Quik® (6 × 1 s dips); (c) tape is dipped into blue stain of
Diff-Quik® (6 × 1 s dips); (d) stain is rinsed off tape under a gentle stream of water; (e) tape is
uncurled by grasping free edge with forceps and laid flat on the glass slide; (f) tape is dried and
flattened firmly onto the glass slide by wiping the surface firmly with a tissue
Interpretation of Cytology Samples Bacteria are very sparse in an oil immersion field (OIF: 1000x magnification). Oil immersion is required for accurate
recognition of bacteria on normal skin surface samples despite being readily
culturable from skin surface swabs (which sample thousands of OIF). The presence of increased numbers of bacteria clustered (colonising) on keratinocytes
represents bacterial overgrowth (Fig. 18), while bacteria present intracellularly
or closely associated within neutrophils (Figs. 19 and 20) and/or macrophages
confirm infection. In deeper samples (e.g. FNA), bacteria should be absent if
VetBooks.ir
236
L. J. Vogelnest
Fig. 18 Numerous cocci
clustered on keratinocytes
on an adhesive tape
impression confirm
bacterial overgrowth, while
cocci intracellularly within
one intact neutrophil
(multi-lobed nucleus)
suggest concurrent focal
bacterial infection (100x
lens, oil immersion;
stained with Diff-Quik® as
per Fig. 17)
Fig. 19 Cocci
intracellularly and
associated with degenerate
neutrophil remnants and
nuclear streaming on an
adhesive tape impression
confirm bacterial infection
(100x lens, oil immersion;
stained with Diff-Quik® as
per Fig. 17)
sterile technique was successfully employed; the presence of any bacteria is
abnormal. Adhesive tape impressions require some experience for efficient and
accurate examination. Keratinocytes typically dominate, staining pale to midblue and ranging from sheets of flat polyhedral cells to single or clustered shards
(follicular cells). Inflammatory cells stain purple, with neutrophils most prevalent; they may be in small clusters or form peripheral rims around keratinocyte
sheets. Eosinophils may also be present, particularly in cases with underlying
hypersensitivity. Neutrophils should be plentiful in erosive or ulcerative samples but may be relatively sparse in drier lesions. Neutrophils degenerate quickly
on the skin surface, often appearing as elongated strands of nuclear material
(nuclear streaming). Tapes should be scanned under low power microscopy (4x
lens) for areas of dense cells or neutrophil clusters to e­ xamine under higher
power (see Fig. 20). Microscope oil is placed directly on the tape surface to
examine under OIF.
VetBooks.ir
Bacterial Diseases
237
Fig. 20 Keratinocytes
distributed singly and in
sheets on an adhesive tape
impression with a central
neutrophil cluster (4x lens;
stained with Diff-Quik® as
per Fig. 17)
Bacterial Culture
Culture and antibiotic susceptibility testing (C&S) is vital for bacterial infections
caused by species with unpredictable antimicrobial sensitivity profiles, such as rods
and many of the environmental bacteria that cause sporadic deep infections. In contrast, empirical therapy based on cytology is considered appropriate for many cases
of SBP [22]. C&S is indicated in severe life-­threatening infections, if rod-shaped
bacteria are evident on cytology (where sensitivity is less predictable), if empirical
therapy does not resolve lesions or when antibiotic resistance is more likely in that
geographical region or patient [1, 22]. There is no current evidence to support any
negative influence of current antibiotic therapy on isolation of causative bacteria;
thus, withdrawal of systemic or topical antibiotics is considered unnecessary [23].
Superficial Skin Sampling Collection of culture samples from primary lesions is
optimal with pustules ruptured and papules incised with a needle prior to sampling
with a culture swab, without preceding skin disinfection [22, 23]. Sterile tissue
biopsy may be more reliable for papules [23]. In dogs with SBP, dry culture swabs
were equally effective as moistened swabs or light scrapings for sampling a range
of superficial lesions, including papules. Swabs were rubbed vigorously for
5–10 seconds on representative lesions, confirmed as SBP on cytology, without
prior skin disinfection [54]. Culture swabs from the skin surface have also been well
utilised for numerous feline skin culture studies sampling a range of skin lesions [5,
7, 9, 17, 19]. Swabs should be immediately placed in transport medium and
VetBooks.ir
238
L. J. Vogelnest
o­ ptimally refrigerated prior to transit to limit overgrowth of contaminants, particularly in warm climates.
Multiple strains of S. pseudintermedius, with distinct antimicrobial resistance profiles, have recently been detected from single lesions in canine SBP,
with pustules and, to a lesser extent, papules associated with less species and
strain diversity than collarettes and crusts. Pustules and papules were swabbed
after incision with the tip of a sterile needle. Crusts and collarettes were sampled by touching a culture swab to the edges of lesions [55]. These findings
reinforce the value of sampling primary lesions whenever possible and raise the
potential importance of collecting multiple samples from a range of primary
lesions to aid identification of all potential pathogens collectively contributing
to infection in a patient.
Deeper Skin Sampling FNA or tissue biopsies collected with sterile technique
are appropriate for bacterial culture from nodular lesions, with tissue samples
most reliable. The surface epidermis may be excised after sample collection to
help avoid isolation of contaminants. Swabs of discharging tracts are not suitable, as a range of contaminant bacteria are readily isolated [22]. When an infectious cause remains uncertain, and a range of infectious agents with varying
culture requirements are differentials, tissue culture samples can be held refrigerated in a sterile container on a sterile saline-moistened swab, pending
histopathology.
Culture Techniques Minimum microbiology evaluation should include complete
speciation of staphylococci, regardless of tube coagulase status, and an antibiogram
for all cultured isolates [1]. In-house culturing can be clinically misleading, resulting in erroneous and ineffective treatments and is not recommended, particularly for
superficial skin sampling [28].
Culture Interpretation Culture results should always be interpreted in light of concurrent cytology findings and the likely pathogens in that location. Growth of bacteria in the laboratory alone does not confirm a pathogenic role. The morphology of
cultured isolates must be consistent with morphology of bacteria evident on cytology
for isolates to be relevant. Even bacteria with alarming multidrug resistance profiles
can be inadvertent contaminants or incidental commensals, without any role in the
current skin disease [1, 22]. However, correctly discerning the relevance of cultured
isolates is not always straightforward; although CoPS are proposed as the major skin
pathogens, commensal CoNS and a variety of environmental saprophytes may be
pathogenic at times, particularly with concurrent immunosuppression [1, 22].
Histopathology
Skin biopsies for histopathology are essential to confirm a diagnosis for many deep
nodular lesions. Multiple excisional biopsies are optimal, sampling any smaller
VetBooks.ir
Bacterial Diseases
239
peripheral lesions in addition to large lesions and avoiding central areas of large
lesions which may be necrotic. Larger lesions should be sectioned to ensure adequate formalin penetration. Biopsies for histopathology should be placed in formalin immediately after collection. Biopsy samples can also be retained frozen for
potential PCR or other molecular testing.
Histopathology is less often indicated for superficial infections but may be
important where cytology results are inconclusive or presentations are atypical for
SBP. Punch biopsies are suitable for small lesions (pustules, papules) or uniform
lesions (plaques, erythema, crusting). Elliptical samples are most useful for transitional areas and edges of ulcerative lesions.
PCR Testing
PCR testing can be helpful to identify species not readily culturable in the laboratory. It is ideally performed on fresh tissue biopsies, collected with sterile technique,
but can also be performed on formalin-fixed samples, assuming fixation in formalin
was for <24 hour. PCR detection from swab samples does not confirm any role as a
pathogen for environmental bacteria (e.g. Nocardia spp.) as detection may simply
reflect skin contaminants.
reatment Principles for Feline Cutaneous Bacterial Infections
T
and Antimicrobial Stewardship
Antimicrobial Resistance and Stewardship
Increasing development of antimicrobial resistance is of profound concern in
recent years and has marked impact on human and animal health and related economics. It is undeniable that antimicrobial use can result in antimicrobial resistance in the species that is being treated and that some resistant pathogens or
resistance mechanisms can be transmitted bi-directionally between animals and
humans [1, 28, 56].
Methicillin resistance of Staphylococcus spp. relevant to veterinary medicine has
been recognised as a serious problem worldwide since the late 1990s, with geographical variation in incidence, but rapid escalation of resistant S. pseudintermedius (MRSP), S. aureus (MRSA) and S. schleiferi species. Acquisition of methicillin
resistance confers resistance to all β-lactam antibiotics, including cephalosporins.
MRS isolates also frequently acquire co-resistance to other classes of antibiotics,
especially FQ and macrolides [18, 19]. MRSP in particular is not uncommonly
multidrug resistant (resistance to at least six antibiotic classes). As S. pseudintermedius is a major canine pathogen and a recognised feline pathogen, this has created
significant new veterinary challenges [1].
Inappropriate use of antibiotics in the veterinary arena is considered an important
factor promoting progression of resistance [1, 28, 56].
VetBooks.ir
240
L. J. Vogelnest
• Cefovecin: Despite being reported as the most frequently chosen antibiotic for
use in cats in recent studies, and specifically the most frequently used for skin
infections or abscesses, it is a third-generation cephalosporin, which is considered ‘highest priority/critically important antimicrobials’ in human medicine,
reserved for life-threatening infections or when culture and susceptibility testing does not indicate alternate antibiotic choices [26, 31]. Reported use is often
‘just in case’, without any clinical and/or cytological evidence to confirm a role
for bacterial infection [31]. Alarmingly, only 0.4% of prescriptions in >1000
cats had C&S testing performed at the time of use and none prior to use. In addition, nearly 23% had concurrent glucocorticoid treatment, with long-acting
methyl-­prednisolone acetate injections in 38% of these, although these drugs
are contraindicated in the face of active infections [31]. Prescription of cefovecin due to convenience of administration is not a justification for valid use.
• Fluoroquinolones: There is evidence that FQ therapy can promote colonisation
with bacteria carrying more resistance genes. FQ therapy was a significant risk
factor for isolation of MRS, multidrug-resistant staphylococci, and FQ-resistant
staphylococci from mucosal samples in dogs in a recent study in England [56].
Clindamycin and amoxi-clav therapy were not significantly associated with
detection of antibiotic resistance, but cephalexin was, potentially due to longer
treatment courses typically used in contrast to amoxi-clav. FQ maintained this
effect at 1 month post-treatment and cephalexin until at least 3 months post-­
treatment [56]. FQ should not be used as first-line treatment options.
Feline MRS Infections There are increasing reports of MRSP and MRSA skin
isolates from cats with skin lesions, although rarely with confirmed pyoderma, in
multiple regions of the world [6, 8, 10, 57]. Variable co-resistance of isolates is
documented, including MRSA with FQ resistance (11.8%) in Australia [10], MRSP
with multidrug resistance in Thailand [57] and MRSP also resistant to TMS (30.8%),
chloramphenicol (7.7%) or clindamycin (7.7%) in Australia [10]. MRSP isolates
from cats are typically sensitive to rifampicin, FQ (second- or third-generation) and
amikacin. CoNS that are more frequently isolated in cats are also often methicillin-­
resistant and multidrug resistant [6].
Risk factors increasing the likelihood of MRS infections in cats are currently
unknown. Risk factors identified in dogs include prior antibiotic therapy, eating
animal stools and contact with veterinary hospitals. Despite confirmed sharing of
staphylococcal isolates including MRSP between pets, dogs from multidog households appear less likely to have mucosal MRS [56].
Antimicrobial Stewardship The appropriate use of antimicrobials to reduce promotion of further antimicrobial resistance is an important concept referred to as
antimicrobial stewardship. The first important principle of appropriate antibiotic
usage is to prescribe antibiotics only in patients with sufficient evidence to confirm
a diagnosis of bacterial infection. Use of antibiotics ‘just in case’, especially without prior diagnostics or when diagnostics fail to confirm bacterial infection, is
strongly discouraged [23, 24, 26, 30, 31].
VetBooks.ir
Bacterial Diseases
241
The second important principle of appropriate antibiotic usage is wise choice of
antibiotic, based on the likely causal bacteria and their likely sensitivity profiles.
Empirical choice is appropriate for diseases where causal pathogens are fairly predictable and have fairly predictable antibiotic susceptibility profiles and first-line
antibiotics (see later) are appropriate. Use of antibiotics that have greater value for
some resistant bacteria (second- or third-line antibiotics) is not suitable without
evidence from C&S that they are appropriate and first-line choices are not, unless
facing life-threatening situations.
The final important principle of appropriate antibiotic usage is to use the correct dose and duration of the chosen antibiotic, taking care to weigh patients accurately prior to therapy and rounding doses up rather than under-dosing (see Table 2).
Although sound evidence is lacking, it is generally recommended that treatment of
superficial infections continues for 3 weeks and deep infections for at least 4 weeks
(and sometimes many months for difficult pathogens). See specific diseases for further guidelines.
Antibiotic Choices
Antibiotic classes are divided into generations based on differences in their spectrum of activity [30], and they can also be divided into groups based on current
prescribing guidelines. There is no clear consensus on optimal antibiotic choices
for bacterial infections in either dogs or cats [1, 26, 28, 30, 31, 58], with a general
paucity of scientific evidence to clarify. The following recommendations for feline
cutaneous bacterial infections are based on a compilation of current expert opinion
in both veterinary and human medicine.
First-line antibiotics are considered most appropriate for empirical therapy
of diagnosed infections, as they are generally well-tolerated and have high efficacy against the expected causal bacteria [26]. Empirical therapy appears suitable
for treatment of feline pyoderma. First-line choices for feline pyoderma are the
following:
• Amoxi-clav or cephalexin – both reported with high levels of sensitivity to isolated Staphylococcus spp. [8] Even in regions where MRS are common in canine
SBP, MRS infections in cats appear very rare, and most reports are of laboratory
isolates [18, 19].
Second-line antibiotics should only be used when there is culture evidence that
first-line drugs will not be effective or as initial empirical therapy for severe infections while awaiting C&S results if resistance to first-line drugs is more likely. This
classification includes newer broad-spectrum antibiotics important to animal and
human health, so reserving their use to necessary cases is prudent. Not all second-­
line choices are equal, with a hierarchical consideration recommended, guided by
regional data [30]. Second-line antibiotics relevant to treatment of feline skin infections include the following:
AMC
(20 –
25
BID)
S b,c
Sb
M
CX (20– DXY a (5 BID)
25 BID)
A
(DBP
only)
S
METR
(10
BID)
GIT
(mild)
GIT
(more)
Oesophageal
stricture (water-swallow)
GIT
(mild)
Sd
Retinal degeneration (enro,
higher doses)
R
R
Myelosuppression;
aplastic anaemia in
people handling
Some MSSP/MSSA
Some MRSP/MRSA
Blood
dyscrasia
M
Second-line: only when C&S supports use and
first-line not suitable; or while C&S pending if
resistance likely (dose mg/kg, frequency)
FQ 2nd Marbo
CHL (50 BID)
TMS (15
CLI
BID)
(5.5–
(2.7–5.5 SID),
11
Enro
BID)
(5 SID)
MSSP/
MSSA
only
Retinal degeneration (orbi,
higher doses)
R
Some MRSP/
MRSA
Severe
risk:
renal,
hepatic,
ototoxic
M
MRSP/
MRSA
Third-line:only when C&S supports
use and no other choices
(dose mg/kg, frequency)
GNT,
CFV e
FQ 3 rd Prado
(8 q
(7.5 SID) Orbi AMK, RIF
14d)
(2.5–7.5 SID)
Critical: (no
veterinary
use)
VAN, TEI,
TEL, LIN
∗
Some regional variation acceptable: judicious antibiotic use requires consideration of local availability, veterinary licensing, recommendations for human use,
and regional antimicrobial susceptibility data [1]
Antibiotic abbreviations: AMC amoxicillin-clavulanic acid, AMK amikacin, CFV cefovecin, CHL chloramphenicol, CLI clindamycin, CX cephalexin, cefadroxil;
d day, DXY doxycycline, Enro enrofloxacin, FQ 2nd second generation fluoroquinolone, FQ 3rd third generation fluoroquinolone, GNT gentamicin, LIN linezolid, Marbo marbofloxacin, Orbi orbifloxacin, Prado pradofloxacin, q every, RIF rifampicin, TEI teicoplanin, TEL telavancin, TMS trimethoprim sulphonamide, VAN vancomycin
Side
effects
Abscess
S
M
M
/cellulitis
Nocardia
R
R
M
R
Rhodococcus
R
R
M
R
No antibiotics “Just in case” use strongly
Uncertain
discouraged[24,56]
SBP/ DBP
Diagnosis
First-line: potential empirical therapy
(dose mg/kg, frequency)
Table 2 Systemic antibiotic choices for feline bacterial infections in line with antimicrobial stewardship guidelines∗ [26, 28, 30, 31, 58]
VetBooks.ir
242
L. J. Vogelnest
General abbreviations: A potential adjunctive value only: not as sole treatment, C&S culture and antibiotic susceptibility testing, DBP deep bacterial pyoderma,
GIT gastro-intestinal tract, M some resistant isolates, at least in some geographical regions, MSSP methicillin-sensitive Staphylococcus pseudintermedius,
MRSP methicillin-resistant Staphylococcus pseudintermedius, MSSA methicillin-­sensitive Staphylococcus aureus, MRSA methicillin-resistant Staphylococcus
aureus, MSSA methicillin-sensitive Staphylococcus aureus, R high levels of resistance for common causal bacteria, S typically high levels of sensitivity for
causal bacteria, SBP superficial bacterial pyoderma
a
May be best considered 2nd-line, particularly in regions where MRSP-isolates are more often susceptible to doxycycline; Minocyclin 8mg/kg once daily can
be used if doxycycline unavailable/expensive
b
Assuming intracellular cocci are present on cytology
c
May be the choice when cocci and rods are present on cytology; C&S is indicated if rods are exclusively present on cytology
d
Resistance occurs with some Bacteroides spp,. and most gram-negative bacteria
e
Often considered second-line, or even first-line; however, third-generation cephalosporins are considered third-line in human medicine
VetBooks.ir
Bacterial Diseases
243
VetBooks.ir
244
L. J. Vogelnest
• Clindamycin – registered for use in many countries for skin and soft-tissue
infections. Although there is some debate in veterinary medicine, macrolide
antibiotics are not first-line choices in human medicine [30]. Clindamycin has
also been shown to have lower levels of sensitivity to staphylococcal isolates in
some studies, and a bacterial culture and susceptibility test is recommended
prior to its use [8].
• Doxycycline – considered first-line in some regions. However, it may be generally less suitable as a first-line choice considering that high levels of resistance
are documented in staphylococcal isolates in some regions [10, 29], even though
lower resistance in others [8]. Minocycline has a similar spectrum of action to
doxycycline and is less expensive and more available in some countries but may
be associated with more gastrointestinal irritation [30].
• Cefovecin – effective against some gram-negative and anaerobic bacteria in addition to gram-positive bacteria, providing a broader spectrum of activity than
second-­generation cephalosporins such as cephalexin. There is generally poor
activity against Pseudomonas spp. and enterococci. Although typically considered first- or second-line in veterinary medicine, third-generation cephalosporins
are considered critically important antibiotics in human medicine reserved for
life-threatening diseases (third-line), so classification as second-line is questioned [30].
• Second-generation FQ (enrofloxacin, difloxacin, marbofloxacin, ciprofloxacin) – primarily target gram-negative bacteria, which are less frequent skin
pathogens.
• Trimethoprim-sulphonamides – greater risk of side effects in cats and lower sensitivity of many bacteria compared to other choices reduce the suitability of this
option; may be effective for some MRS.
Third-line antibiotics are very important to animal and human health, especially for treatment of multidrug-resistant bacteria, and their use should be only
considered when C&S indicates a lack of other treatment choices. Many are not
licensed for veterinary use [26, 30]. Their use for superficial infections is strongly
discouraged. Third-line choices for cats with severe bacterial cutaneous infections
include the following:
• Third-generation FQ (pradofloxacin and orbifloxacin) – have an increased
gram-­positive and anaerobic spectrum compared to second-generation FQ, in
addition to good gram-negative coverage; considered unlikely to be effective
for Nocardia spp. [30].
• Aminoglycosides (gentamicin, amikacin) – potential considerations only for life-­
threatening skin infections, but have considerable risk of severe renal side effects,
requiring careful monitoring, concurrent fluid therapy and brief duration
therapy
VetBooks.ir
Bacterial Diseases
245
• Other new and old antibiotics (chloramphenicol, clarithromycin, rifampicin,
imipenem, piperacillin) – potential use for MRS and multidrug-resistant bacteria, but considerable potential for moderate to severe side effects
• Newest generation antibiotics (e.g. vancomycin, teicoplanin, telavancin, linezolid) – deemed of critical importance to human health and strongly discouraged/unavailable for veterinary use [1, 26]
Management of Veterinary Patients with MRS Infection
Transmission of MRS between humans and various animal species including cats
is documented [1, 28]. MRSA and methicillin-resistant CoNS, including S. haemolyticus, S. epidermidis and S. fleurettii, were co-isolated from multiple cats,
horses and humans on one farm in Europe, with isolates sharing the same characteristics [59]. Concern is thus raised when MRS infections are documented in
veterinary species, when greater bacterial numbers are likely to increase the risks
of transmission.
It is currently recommended that pets with MRS infections have limited contact with other pets or humans until their infections are controlled and that good
hand hygiene and heightened cleaning protocols are used in the home environment to reduce potential transmission. Veterinary hospitals are also recognised
as potential sources of MRS transmission, and adherence to strict hand hygiene
(proper washing/drying and use of alcohol-based hand sanitizers) between handling all patients and regular cleaning and disinfection protocols will reduce the
risks of transmission, with MRS susceptible to commonly used disinfectants.
Barrier nursing protocols for hospitalised patients with known MRS infections
are recommended [1, 56].
Despite concerns over the potential challenges of treatment of MRS infection,
resistant isolates are not more virulent or likely to cause infection than non-resistant isolates. There is no current evidence to support attempted decolonisation of
patients colonised by MRS, and thus, screening of clinically normal animals for
carriage of MRS is currently not recommended [1].
Conclusion
Feline cutaneous bacterial infections range from common secondary to rare but
potentially life-threatening deep and disseminated infections. Causal pathogens
include normal skin and mucosal commensals and a range of environmental saprophytes. Development of antimicrobial resistance, particularly methicillin resistance
in staphylococci, poses increasing veterinary challenges. Accurate and efficient
VetBooks.ir
246
L. J. Vogelnest
diagnosis is important to expedite appropriate treatment and to limit further promotion of antibiotic resistance by restricting use of antibiotics to patients with confirmed disease.
References
1. Morris DO, Loeffler A, Davis MF, Guardabassi L, Weese JS. Recommendations for approaches
to methicillin-resistant staphylococcal infections of small animals: diagnosis, therapeutic considerations and preventative measures: Clinical Consensus Guidelines of the World Association
for Veterinary Dermatology. Vet Dermatol. 2017;28:304–30.
2. Rossi CC, da Silva DI, Mansur Muniz I, Lilenbaum W, Giambiagi-deMarval M. The oral
microbiota of domestic cats harbors a wide variety of Staphylococcus species with zoonotic
potential. Vet Microbiol. 2017;201:136–40.
3. Weese JS. The canine and feline skin microbiome in health and disease. Vet Dermatol.
2013;24:137–45.
4. Patel A, Lloyd DH, Lamport AI. Antimicrobial resistance of feline staphylococci in South-­
Eastern England. Vet Dermatol. 1999;10:257–61.
5. Patel A, Lloyd DH, Howell SA, Noble WC. Investigation into the potential pathogenicity of
Staphylococcus felis in a cat. Vet Rec. 2002;150:668–9.
6. Muniz IM, Penna B, Lilenbaum W. Methicillin-resistant commensal staphylococci in the oral
cavity of healthy cats: a reservoir of methicillin resistance. Vet Rec. 2013;173:502.2. https://
doi.org/10.1136/vr.101971.
7. Igimi SI, Atobe H, Tohya Y, Inoue A, Takahashi E, Knoishi S. Characterization of the most
frequently encountered Staphylococcus sp. in cats. Vet Microbiol. 1994;39:255–60.
8. Qekwana DN, Sebola D, Oguttu JW, Odoi A. Antimicrobial resistance patterns of
Staphylococcus species isolated from cats presented at a veterinary academic hospital in South
Africa. BMC Vet Res. 2017;13:286. https://doi.org/10.1186/s12917-017-1204-3.
9. Abraham JK, Morris DO, Griffeth GC, Shofer FS, Rankin SC. Surveillance of healthy cats
and cats with inflammatory skin disease for colonization of the skin by methicillin-resistant
coagulase-positive staphylococci and Staphylococcus schleiferi ssp. schleiferi. Vet Dermatol.
2007;18:252–9.
10. Saputra S, Jordan D, Worthing KA, Norris JM, Wong HS, Abraham R, et al. Antimicrobial
resistance in coagulase-positive staphylococci isolated from companion animals in Australia:
a one year study. PLoS One. 2017;12:e0176379. https://doi.org/10.1371/0176379.
11. Older CE, Diesel A, Patterson AP, Meason-Smith C, Johnson TJ, Mansell J, Suchodolski J,
Hoffmann AR. The feline skin microbiota: the bacteria inhabiting the skin of healthy and
allergic cats. PLoS One. 2017;12:e0178555. https://doi.org/10.1371/vr.0178555.
12. Wildermuth BE, Griffin CE, Rosenkrantz WS. Feline pyoderma therapy. Clin Tech Small
Anim Pract. 2006;21:150–6.
13. Scott DW, Miller WH, Erb HN. Feline dermatology at Cornell University: 1407 cases (1988–
2003). J Fel Med Surg. 2013;15:307–16.
14. Yu HW, Vogelnest LJ. Feline superficial pyoderma: a retrospective study of 52 cases (2001–
2011). Vet Dermatol. 2012;23:448–55.
15. Whyte A, Gracia A, Bonastre C, Tejedor MT, Whyte J, Monteagudo LV, Simon C. Oral disease
and microbiota in free-roaming cats. Top Companion Anim Med. 2017;32:91–5.
16. Wooley KL, Kelly RF, Fazakerley J, Williams NJ, Nuttal TJ, McEwan NA. Reduced in vitro
adherence of Staphylococcus spp. to feline corneocytes compared to canine and human corneocytes. Vet Dermatol. 2006;19:1–6.
17. Medleau L, Blue JL. Frequency and antimicrobial susceptibility of Staphylococcus spp isolated from feline skin lesions. J Am Vet Med Assoc. 1988;193:1080–1.
VetBooks.ir
Bacterial Diseases
247
18. Morris DO, Rook KA, Shofer FS, Rankin SC. Screening of Staphylococcus aureus,
Staphylococcus intermedius, and Staphylococcus schleiferi isolates obtained from small companion animals for antimicrobial resistance: a retrospective review of 749 isolates (2003–04).
Vet Dermatol. 2006;17:332–7.
19. Morris DO, Maudlin EA, O’Shea K, Shofer FS, Rankin SC. Clinical, microbiological, and
molecular characterization of methicillin-resistant Staphylococcus aureus infections of cats.
Am J Vet Res. 2006;67:1421–5.
20. Selvaraj P, Senthil KK. Feline Pyoderma – a study of microbial population and its antibiogram.
Intas Polivet. 2013;14(11):405–6.
21. White SD. Pyoderma in five cats. J Am Anim Hosp Assoc. 1991;27:141–6.
22. Beco L, Guaguere E, Lorente Mendez C, Noli C, Nuttall T, Vroom M. Suggested guidelines
for using systemic antimicrobials in bacterial skin infections (1): diagnosis based on clinical
presentation, cytology and culture. Vet Rec. 2013;172:72–8.
23. Hillier A, Lloyd DH, Weese JS, Blondeau JM, Boothe D, Breitschwerdt E, et al. Guidelines
for the diagnosis and antimicrobial therapy of canine superficial bacterial folliculitis
(Antimicrobial Guidelines Working Group of the International Society for Companion Animal
Infectious Diseases). Vet Dermatol. 2014;25:163–74.
24. Singleton DA, Sanchez-Vizcaino F, Dawson S, Jones PH, Noble PJ, Pinchbeck GL, et al.
Patterns of antimicrobial agent prescription in a sentinel population of canine and feline veterinary practices in the United Kingdom. The Vet J. 2017;224:18–24.
25. Wildermuth BE, Griffin CE, Rosenkrantz WS. Response of feline eosinophilic plaques and lip
ulcers to amoxicillin trihydrate–clavulanate potassium therapy: a randomized, double-blind
placebo-controlled prospective study. Vet Dermatol. 2011;23:110–8.
26. Beco L, Guaguere E, Lorente Mendez C, Noli C, Nuttall T, Vroom M. Suggested guidelines
for using systemic antimicrobials in bacterial skin infections (2): antimicrobial choice, treatment regimens and compliance. Vet Rec. 2013;172:156–60.
27. Borio S, Colombo S, La Rosa G, De Lucia M, Dombord P, Guardabassi L. Effectiveness of a
combined (4% chlorhexidine digluconate shampoo and solution) protocol in MRS and non-­
MRS canine superficial pyoderma: a randomized, blinded, antibiotic-controlled study. Vet
Dermatol. 2015;26:339–44.
28. Weese JS, Giguere S, Guardabassi L, Morley PS, Papich M, Ricciuto DR, et al. ACVIM consensus statement on therapeutic antimicrobial use in animals and antimicrobial resistance. J
Vet Intern Med. 2015;29:487–98.
29. Mohamed MA, Abdul-Aziz S, Dhaliwal GK, Bejo SK, Goni MD, Bitrus AA, et al. Antibiotic
resistance profiles of Staphylococcus pseudintermedius isolated from dogs and cats. Malays J
Microbiol. 2017;13:180–6.
30. Whitehouse W, Viviano K. Update in feline therapeutics: clinical use of 10 emerging therapies.
J Feline Med Surg. 2015;17:220–34.
31. Burke S, Black V, Sanchez-Vizcaino F, Radford A, Hibbert A, Tasker S. Use of cefovecin
in a UK population of cats attending first-opinion practices as recorded in electronic health
records. J Feline Med Surg. 2017;19:687–92.
32. Hardefeldt LY, Holloway S, Trott DJ, Shipstone M, Barrs VR, Malik R, et al. Antimicrobial
prescribing in dogs and cats in Australia: results of the Australasian Infectious Disease
Advisory Panel Survey. J Vet Intern Med. 2017;31:1100–7.
33. Scott DW, Miller WH. Feline acne: a retrospective study of 74 cases (1988–2003). Jpn J Vet
Dermatol. 2010;16:203–9.
34. Jazic E, Coyner KS, Loeffler DG, Lewis TP. An evaluation of the clinical, cytological, infectious and histopathological features of feline acne. Vet Dermatol. 2006;17:134–40.
35. Walton DK, Scott DW, Manning TO. Cutaneous bacterial granuloma (botryomycosis) in a dog
and cat. J Am Anim Hosp Assoc. 1983;183(19):537–41.
36. Murai T, Yasuno K, Shirota K. Bacterial pseudomycetoma (Botryomycosis) in an FIV-positive
cat. Jap J Vet Dermatol. 2010;16:61–5.
VetBooks.ir
248
L. J. Vogelnest
37. Norris JM, Love DN. The isolation and enumeration of three feline oral Porphyromonas species from subcutaneous abscessed in cats. Vet Microbiol. 1999;65:115–22.
38. Traslavina RP, Reilly CM, Vasireddy R, Samitz EM, Stepnik CT, Outerbridge C, et al. Laser
capture microdissection of feline Streptomyces spp pyogranulomatous dermatitis and cellulitis. Vet Pathol. 2015;205(52):1172–5.
39. De Araujo FS, Braga JF, Moreira MV, Silva VC, Souza EF, Pereira LC, et al. Splendore-­
Hoeppli phenomenon in a cat with osteomyelitis caused by Streptococcus species. J Feline
Med Surg. 2014;16:189–93.
40. Malik R, Krockenberger MB, O’Brien CR, White JD, Foster D, Tisdall PL, et al. Nocardia
infections in cats: a retrospective multi-institutional study of 17 cases. Aust Vet J. 2006;84:
235–45.
41. Gunew MN. Rhodococcus equi infection in cats. Aust Vet Practit. 2002;32:2–5.
42. Farias MR, Takai S, Ribeiro MG, Fabris VE, Franco SR. Cutaneous pyogranuloma in a
cat caused by virulent Rhodococcus equi containing an 87 kb type I plasmid. Aust Vet J.
2007;85:29–31.
43. Patel A. Pyogranulomatous skin disease and cellulitis in a cat caused by Rhodococcus equi. J
Small Anim Pract. 2002;43:129–32.
44. Miller RI, Ladds PW, Mudie A, Hayes DP, Trueman KF. Probable dermatophilosis in 2 cats.
Aust Vet J. 1983;60:155–6.
45. Kaya O, Kirkan S, Unal B. Isolation of Dermatophilus congolensis from a cat. J Veterinary
Med Ser B. 2000;47:155–7.
46. Carakostas MC. Subcutaneous dermatophilosis in a cat. J Am Vet Med Assoc.
1984;185:675–6.
47. Sharman MJ, Goh CS, Kuipers RG, Hodgson JL. Intra-abdominal actinomycetoma in a cat. J
Feline Med Surg. 2009;11:701–5.
48. Koenhemsi L, Sigirci BD, Bayrakal A, Metiner K, Gonul R, Ozgur NY. Actinomyces viscosus
isolation from the skin of a cat. Isr J Vet Med. 2014;69:239–42.
49. Kruger EF, Byrne BA, Pesavento P, Hurley KF, Lindsay LL, Sykes JE. Relationship between
clinical manifestations and pulsed-field gel profiles of Streptococcus canis isolates from dogs
and cats. Vet Microbiol. 2010;146:167–71.
50. Nolff MC, Meyer-Lindenberg A. Necrotising fasciitis in a domestic shorthair cat – negative pressure wound therapy assisted debridement and reconstruction. J Small Anim Pract.
2015;56:281–4.
51. Brachelente C, Wiener D, Malik Y, Huessy D. A case of necrotizing fasciitis with septic shock
in a cat caused by Acinetobacter baumannii. Vet Dermatol. 2007;18:432–8.
52. Plavec T, Zdovc I, Juntes P, Svara T, Ambrozic-Avgustin I, Suhadolc-Scholten S. Necrotising
fasciitis, a potential threat following conservative treatment of a leucopenic cat: a case report.
Vet Med (Praha). 2015;8:460–7.
53. Berube DE, Whelan MF, Tater KC, Bracker KE. Fournier’s gangrene in a cat. J Vet Emerg Crit
Care. 2010;20:148–4.
54. Ravens PA, Vogelnest LJ, Ewen E, Bosward KL, Norris JM. Canine superficial bacterial pyoderma: evaluation of skin surface sampling methods and antimicrobial susceptibility of causal
Staphylococcus isolates. Aust Vet J. 2014;92:149–55.
55. Larsen RF, Boysen L, Jessen LR, Guardabassi L, Damborg P. Diversity of Staphylococcus
pseudintermedius in carriage sites and skin lesions of dogs with superficial bacterial folliculitis: potential implications for diagnostic testing and therapy. Vet Dermatol.
2018;29:291–5.
56. Schmidt VM, Pinchbeck G, Nuttall T, Shaw S, McIntyre KM, McEwan N, et al. Impact of
systemic antimicrobial therapy on mucosal staphylococci in a population of dogs in Northwest
England. Vet Dermatol. 2018;29:192–202.
57. Kadlec K, WeiB S, Wendlandt S, Schwarz S, Tonpitak W. Characterization of canine and feline
methicillin-resistant Staphylococcus pseudintermedius (MRSP) from Thailand. Vet Microbiol.
2016;194:93–7.
VetBooks.ir
Bacterial Diseases
249
58. Lappin MR, Bondeau J, Boothe D, Breitschwerdt FB, Guardabassi L, Lloyd DH, et al.
Antimicrobial use Guidelines for Treatment of Respiratory Tract Disease in Dogs and Cats:
Antimicrobial Guidelines Working Group of the International Society for Companion Animal
Infectious Diseases. J Vet Intern Med. 2017;31:279–94.
59. Loncaric I, Kunzel F, Klang A, Wagner R, Licka T, Grunert T, et al. Carriage of methicillin-­
resistant staphylococci between humans and animals on a small farm. Vet Dermatol.
2016;27:191–4.
VetBooks.ir
Mycobacterial Diseases
Carolyn O’Brien
Abstract
Cats may be infected with a variety of both rapidly- and slowly-growing mycobacterial species, which cause a variety of clinical syndromes in cats, from localized skin disease to disseminated and potentially fatal infections. Cutaneous
disease is the most common manifestation for all causative species; however,
some species may have internal involvement, with any organ system, skeletal or
soft tissue structure potentially infected. Infections by rapidly-growing mycobacteria generally result in fistulating panniculitis of the inguinal region or less
commonly, axillae, flanks or dorsum, whereas those caused by members of the
slow-growing taxons typically present with solitary or multiple nodular skin
lesions and/or local lymphadenopathy, especially of the head, neck and/or limbs.
Most affected cats do not appear to have an underlying immunosuppressive condition, and no association has been made with a positive retroviral status. Most
cases occur in adult cats with unrestricted outdoor access. Depending on the
causative species and the extent of disease when first diagnosed, these infections
can be challenging to treat. Generally, localized cutaneous infection caused by
all species has a relatively favorable prognosis if treated with an appropriate
combination of drugs and surgery, if necessary. If the cat acquires systemic infection, the prognosis becomes significantly worse. The commitment of the owner
to the implementation of a potentially expensive and time-consuming schedule
of multidrug therapy for many months may also influence the outcome. The zoonotic potential of these organisms is generally low, however cat-to-human transfer of Mycobacterium bovis has been reported.
C. O’Brien (*)
Melbourne Cat Vets, Fitzroy, Victoria, Australia
e-mail: cob@catvet.net.au
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_12
251
VetBooks.ir
252
C. O’Brien
Mycobacteria are aerobic, nonmotile, Gram-positive, nonspore-forming bacilli in
the phylum Actinobacteria. Of the more than 180 mycobacterial species identified
[1], almost all are environmental saprophytes. However, a few, such as the
Mycobacterium tuberculosis complex (MTB), M. leprae and its relatives, members
of the M. avium complex (MAC), such as M. avium subsp. paratuberculosis and M.
lepraemurium, appear to have evolved into obligate pathogens.
Mycobacterial species can be divided genetically and phenotypically into two
main groups: rapidly growing (RGM) and slowly growing mycobacteria (SGM).
The RGM are ancestral to the SGM, with the latter forming a distinct genetic subbranch based on analysis of housekeeping genes and, more recently, whole-genome
analysis [2]. The M. abscessus/chelonae complex appears to be the genetically oldest group identified, with M. triviale and also the closely related M. terrae group the
likely evolutionary links between the RGM and the SGM [2].
Mycobacterial infections cause a variety of clinical syndromes in cats, from
minor localized skin disease to potentially fatal disseminated infections. Cutaneous
disease is the most common manifestation for all causative species; however, some
species, particularly the MTB and MAC, may have internal involvement with any
organ system, skeletal or soft tissue structure, potentially infected.
Few investigations have examined substantial cohorts of cats with mycobacteriosis and only some definitively identified the causative mycobacterial species via genetic analysis. These studies are typically limited to animals from a
particular geographical region and may not be representative of the disease in
cats domiciled elsewhere, especially with regard to incidence and causative
species.
Typically, cats with mycobacterial infections do not appear to have an immunosuppression, and no association has been made with a positive retroviral status,
unlike MAC infections in people with human immunodeficiency virus/acquired
immunodeficiency syndrome. Regardless of the causative species, most cases occur
in adult cats with unrestricted outdoor access, although MAC infections have been
occasionally reported in exclusively indoor cats.
Rapidly Growing Mycobacteria
Etiology and Epidemiology
The RGM are environmental saprophytes widely distributed as free-living organisms in both terrestrial and aquatic biomes. The RGM are so named as they are able
to grow on synthetic culture media within 7 days at 75–113° F (24–45° C).
RGM have low inherent pathogenicity and generally tend to cause opportunistic
infections in cats, mostly through breaches in the integument, for example, via cat-­
scratch wounds. They have a low tendency to cause systemic disease unless the host
is immunocompromised, although occasionally inhalation of organisms may lead to
pneumonia in apparently immunocompetent individuals. The disease manifests in
cats primarily as ventral abdominal panniculitis and tends to be caused by the M.
smegmatis, M. margaritense, M. fortuitum, and M. chelonae-abscessus groups.
VetBooks.ir
Mycobacterial Diseases
253
Cases are reported from the Americas (Brazil, southeastern and southwestern
United States, Canada), Oceania (Australia and New Zealand), and Europe (Finland,
the Netherlands, Germany, and the United Kingdom). The incidence of particular
causative organisms varies between geographical regions. M. smegmatis and M.
margaritense, followed by M. fortuitum groups, cause most infections in cats in
eastern Australia, whereas, in the southwestern United States, M. fortuitum group
followed by M. chelonae infections appear to be more common.
Cats with a prominent ventral abdominal fat pad appear to have a predisposition
toward RGM infection. This is likely due to the preference of the organisms for tissues rich in lipid, which may provide triglycerides for growth and perhaps protection from the host immune response. In cats that do not have a significant amount of
subcutaneous fat, the ability to establish experimental infections appears to be limited [3].
Clinical Features
Typically, lesions caused by RGM are located in the inguinal region or, less
commonly, axillae, flanks, or dorsum. Initially, the infection appears as a circumscribed plaque or nodule of the skin and subcutis. Subsequently, the affected
cat develops alopecic areas of thin epidermis which overlies and is adherent to
diseased subcutaneous tissue; this results in a characteristic “pepper pot”
appearance (Fig. 1). The characteristic focal purple depressions in the skin
break down to become fistulae exuding a watery discharge that may become
purulent with secondary infection. The lesions may eventually involve the entire
ventral abdomen, flanks, perineum, and occasionally the limbs. Internal organs
or lymph node involvement is not likely; however, the abdominal wall is rarely
involved.
Most cats do not have signs of systemic illness unless the skin lesions become
secondarily infected with Staphylococcus and Streptococcus spp., in which case
the patient may display lethargy, pyrexia, anorexia, weight loss, and reluctance to
move.
Diagnosis
Fine needle aspiration and cytology may establish the presence of pyogranulomatous inflammation, and subcutaneous exudate may be obtained with this technique
to allow the culture of the organism, thus establishing the diagnosis.
RGM are not typically visible on either Romanowsky-stained cytological samples or hematoxylin and eosin-stained histopathology sections of biopsy tissue.
Instead, they are visualized using acid-fast stains, such as Ziehl-Neelsen (ZN) or
Fite’s.
RGM may be few in number and difficult to visualize in acid-fast stained cytological material, and the diagnosis is not excluded if organisms are not visualized.
The organisms may be lost during processing of cytologic and histopathologic
VetBooks.ir
254
C. O’Brien
Fig. 1 The typical
appearance of dermatitis/
panniculitis caused by a
rapidly growing
mycobacterial species,
Mycobacterium smegmatis.
(Courtesy of Nicola
Colombo)
samples as they tend to exist extracellularly in fat vacuoles in tissues. Occasionally,
positive results on mycobacterial culture or molecular methods such as polymerase
chain reaction (PCR) may be obtained on samples that are “acid-fast bacilli (AFB)negative” on cyto- or histopathological evaluation.
Punch biopsies of the skin are usually inadequate for obtaining representative
tissue samples, and a deep subcutaneous tissue biopsy from the margin of the lesion
is preferred. The histopathologic characteristics of RGM dermatitis/panniculitis
include an ulcerated or acanthotic dermis overlying multifocal to diffuse pyogranulomatous inflammation, which tends to extend well into the subcutis. In the pyogranulomas, a rim of neutrophils often surrounds a clear, inner zone of degenerate
adipocytes, which may contain scant AFB with an outer collection of epithelioid
macrophages (Fig. 2). A mixed inflammatory response, predominantly comprising
neutrophils and macrophages, but also containing lymphocytes and plasma cells, is
found between each pyogranuloma. AFB may also occasionally be visualized
within macrophages but can be very hard to find within tissue sections.
When attempting to culture mycobacteria from panniculitis lesions, material
swabbed directly from cutaneous draining sinus tracts usually contains high numbers of contaminating skin bacteria, which outcompete the RGM on culture media.
Mycobacterial Diseases
b
VetBooks.ir
a
255
Fig. 2 (a) Histopathological aspect of rapidly growing mycobacterial infection: pyogranulomatous
inflammation with a rim of neutrophils surrounding a clear, inner zone of degenerate adipocytes,
which contain acid fast bacteria (H&E 400×); (b) Ziehl-Neelsen stain of the same sample: rod
shaped bacteria are stained in red and can be easily recognised (400×). (Courtesy of Dr. Chiara Noli)
Fine-needle samples obtained through intact skin decontaminated with 70% ethanol
or surgically collected subcutaneous tissue biopsies are therefore preferred.
Uncontaminated samples of RGM grow readily on routine media such as blood [4]
and MacConkey agar (without crystal violet), so there is usually no need for the
clinician to request “mycobacterial media” culture for these organisms specifically.
Treatment and Prognosis
Depending on the causative species and the extent of disease when first diagnosed,
these infections can be challenging to treat. They often have a high rate of recurrence, frequently require protracted courses of therapy, and may have a substantial
incidence of inherent and/or acquired drug resistance.
Susceptibility data is especially useful for organisms that may have inherently
variable drug susceptibility, such as M. fortuitum, or for recurrent or chronically
persistent RGM infections, especially where the cat has undergone prior antibiotic
treatment which may have induced acquired drug resistance. Ideally, treatment
should begin with one or two oral antimicrobials (doxycycline, a fluoroquinolone,
and/or clarithromycin). These are usually chosen empirically until results of culture
and susceptibilities are known. In Australia, doxycycline and/or a fluoroquinolone –
preferably pradofloxacin – are best, whereas, in the United States, clarithromycin is
the drug of choice initially. M. smegmatis group tends to be inherently resistant to
clarithromycin, and some isolates may be resistant to the enrofloxacin or ciprofloxacin, although this does not rule out susceptibility to pradofloxacin [5]. Members of
the M. fortuitum group are typically susceptible to fluoroquinolones, however, demonstrate variable expression of the erythromycin-inducible methylase (erm) gene
which confers macrolide resistance [6]. Approximately 50% of M. fortuitum isolates are susceptible to doxycycline [7]. M. chelonae-abscessus group isolates tend
to be resistant to all drugs available for oral dosing apart from clarithromycin and
VetBooks.ir
256
C. O’Brien
linezolid. Where indicated by drug susceptibility data, refractory cases may be
treated with clofazimine, amikacin, cefoxitin, or linezolid. It is recommended to
commence treatment at standard dose rates increased slowly to the high end of the
dose range, unless adverse effects are observed.
Treatment duration is variable, but it is recommended to continue therapy for
1–2 months past resolution of all clinical signs. Some animals with recalcitrant
lesions benefit from en bloc resection of isolated areas of infection, often necessitating reconstructive surgery [8] or vacuum-assisted wound closure [9, 10].
Public Health Risks
Zoonotic transmission of RGM organisms from infected animals to humans is very
unlikely. There is one report of M. fortuitum infection in an otherwise healthy
middle-­aged woman, after a cat bite to the forearm [11].
Slowly Growing Mycobacteria
The SGM taxon includes a large number of opportunistic environmental species:
the obligate pathogens, M. leprae and M. lepromatosis, and the members of the M.
tuberculosis complex. There are also a number of fastidious species included – traditionally classified as the causative species of “feline leprosy” – that are incapable
of growing in axenic culture; thus, their epidemiological niche is unclear.
Tuberculous Mycobacteria
Cats are naturally resistant to M. tuberculosis, but occasional infections likely transmitted directly from humans are reported [12]. Disease in cats is most commonly
caused by M. bovis and M. microti [13]. M. bovis has worldwide endemicity.
However, much of Continental Europe, parts of the Caribbean, and Australia are
free of the disease due to widespread surveillance, slaughter of test-positive cattle,
the pasteurization of milk, and the absence of a wildlife host. M. microti is endemic
to Europe and the United Kingdom (UK). Its main reservoir appears to be voles,
shrews, wood mice, and other small rodents [14].
The exact route of transmission of these MTB species to cats is unclear. Numerous
potential rodent prey species collected from areas of southwest England were found
to be infected with M. bovis [15]. Suspected nosocomial contamination of surgical
wounds has been reported [16].
MAC and Other Slowly Growing Saprophytes
Disease in cats is caused by several saprophytic slowly growing mycobacterial species, mostly members of the MAC, which are found worldwide in water sources and
soil. Certain slowly growing species are more common in some environmental
VetBooks.ir
Mycobacterial Diseases
257
niches or particular geographical areas, for example, M. malmoense or biofilms with
M. intracellulare in the UK and Sweden. Some have highly restricted, focal areas of
endemicity, for example, M. ulcerans infection.
As with MTB complex, the clinical picture is determined by the route of infection. Cats likely acquire skin lesions via transcutaneous inoculation of contaminated
environmental material. Most cats with slow-growing mycobacterial infections
have unrestricted outdoor access, and almost all of these cases had no overt predisposing conditions.
Fastidious Mycobacteria
“Feline leprosy” has been diagnosed in New Zealand, Australia, western Canada,
the UK, southwestern United States, continental Europe, New Caledonia, the Greek
islands, and Japan. Historically, New Zealand and Australia have reported the highest number of cases worldwide.
Genetic studies have identified the involvement of several “non-culturable” species of mycobacteria: M. lepraemurium, Candidatus “M. tarwinense,‘ [17, 18]
Candidatus “M. lepraefelis,‘ [19] and M. visibilis, although the latter has not been
reported for many years [20]. M. lepraemurium tends to cause disease in young
male cats, whereas Candidatus “M. tarwinense” and Candidatus “M. lepraefelis”
are more likely to cause disease in middle-aged to older cats. There is no gender
preponderance for Candidatus “M. tarwinense” infection, whereas Candidatus “M.
lepraefelis” is slightly more likely to cause disease in males.
Clinical Features
The majority of cats with SGM infection have solitary or multiple nodular skin lesions
and/or local lymphadenopathy, especially of the head, neck, and/or limbs (Fig. 3).
Ulceration of cutaneous lesions and the skin overlying affected lymph nodes may be
Fig. 3 Large, ulcerated
nodules on the lateral
thigh of a young male
cat with Mycobacterium
lepraemurium infection.
Despite the widespread
nature of the cutaneous
lesions, this cat was
cured with multidrug
therapy including
rifampicin and
clofazimine. (Courtesy
of Dr. Mei Sae Zhong)
VetBooks.ir
258
C. O’Brien
observed, and infection may occasionally involve contiguous muscle and bone, which
is more often the case with MTB complex species than other causative agents. In
some cases, the dermal lesions may be widespread, involving many cutaneous sites.
Host factors (age, concurrent illness, immunological status), the causal species, or the
route and size of the inoculum may influence the nature of the disease.
If systemic disease is detected, the most common causative agents are either the
MTB complex mycobacteria (especially in the UK and New Zealand) or members of
the MAC. Rarely, systemic infections by other mycobacterial species, including other
slowly growing saprophytes and Candidatus “M. lepraefelis,” have been documented.
Diagnosis
Differential diagnoses of nodular skin and subcutaneous lesions include Nocardia
and Rhodococcus spp. (which may also be acid-fast), fungi, or algal infections and
primary or metastatic neoplasia. There are no pathognomonic clinical features that
differentiate mycobacterial infections from other etiologies, and collection of representative tissue samples for cytology or histopathology and microbiology is necessary for the diagnosis.
It is vital in areas endemic for the MTB complex that the diagnosis is not based
simply on cytologic or histopathologic findings. An attempt to identify the causative
agent should be made in every case, ideally via a mycobacterium reference laboratory or equivalent, especially where mandatory reporting of such cases may result
in compulsory euthanasia.
The diagnosis of cutaneous infections caused by SGM is often relatively simple,
provided there is a high index of suspicion. Personal protective equipment should be
worn during any procedure which involves handling of discharging or ulcerated
lesions and/or surgical or necropsy tissues, when members of the MTB complex are
a possible cause of disease.
Ideally, at the time of biopsy sampling for histopathology, a piece of fresh tissue
wrapped in sterile saline-moistened gauze swabs placed in a sterile container should
be collected, if microbiological processing is needed. The pathology laboratory
should ideally be notified before submission, as SGM culture and identification
requires specialized expertise.
Romanowsky-stained cytological samples of cutaneous nodules will demonstrate granulomatous to pyogranulomatous inflammation, and mycobacteria are
recognized by their characteristic “negatively staining” appearance (Fig. 4), usually located within macrophages. As with the RGM, SGM are not typically visible
on Romanowsky-stained cytological or hematoxylin and eosin-stained histopathology sections, except M. visibile and Candidatus “M. lepraefelis.” Instead,
Ziehl-Neelsen (ZN) staining (Fig. 5) or similar (e.g. Fite’s) is required. Depending
on mycobacterial species and host immune response, bacterial numbers may be
variable.
MTB complex organisms produce characteristic solitary to coalescing granulomas (“tubercules”). Granulation tissue surrounds a layer of mixed inflammatory
cells, consisting of macrophages, neutrophils, lymphocytes, and plasma cells. The
VetBooks.ir
Mycobacterial Diseases
259
Fig. 4 Numerous
macrophages with
cytoplasm filled with many
achromatic rod-shaped
areas (Diff Quick, 1000×).
(Courtesy of Dr. Francesco
Albanese)
Fig. 5 Many brightly red
and rod-shaped
Mycobacteria are well
recognizable with
Ziehl-Neelsen staining
(1000×). (Courtesy of Dr.
Francesco Albanese)
center of the granuloma contains epithelioid macrophages and some neutrophils,
with variable but usually low numbers of AFB, with or without necrotic tissue.
Cutaneous MAC infections cause pyogranulomatous or granulomatous inflammation with a variable fibroblastic response. The fibroblastic reaction may, on occasion, be so pronounced as to make it difficult to differentiate the disease from an
inflamed fibrosarcoma (so-called “mycobacterial pseudotumor”) [21]. AFB found
both within macrophages and spindle cells identify the underlying etiology in these
cases (Fig. 6). In the absence of a prominent fibroblastic response, lesions may
resemble lepromatous leprosy.
The pathological picture of feline leprosy is subdivided into multi-bacillary (lepromatous) and pauci-bacillary (tuberculoid) forms [22]. “Multi-bacillary” leprosy is
thought to correspond with a weak cell-mediated immune (CMI) response. Typically,
many foamy or multinucleate macrophages, containing huge numbers of mycobacteria, are observed. There is no necrosis, and lesions contain virtually no lymphocytes and plasma cells. “Pauci-bacillary” leprosy, in which moderate to few
VetBooks.ir
260
C. O’Brien
Fig. 6 Histopathological
appearance of MTB
complex infection:
granulomas consisting of
macrophages, neutrophils,
lymphocytes and plasma
cells. The center of the
granuloma contains
epithelioid macrophage
(H&E 400×) (Courtesy of
Dr. Chiara Noli)
observable AFB are found within pyogranulomatous inflammation dominated by
epithelioid histiocytes, is thought to occur with a more effective CMI response.
Moderate numbers of lymphocytes and plasma cells are also observed, with multifocal to coalescing necrosis. Involvement of peripheral nerves, a feature of human
leprosy, is not seen in cats.
Except where samples have become contaminated with environmental mycobacteria, molecular methods, such as PCR and sequencing, can provide a highly accurate diagnosis on fresh or frozen tissue, formalin-fixed paraffin-embedded tissue
sections, and Romanowsky-stained cytology slides [23]. It should be remembered
that for samples in which no AFB are visualized microscopically, mycobacterial
infections cannot be excluded with a negative PCR result.
A feline IFN-γ ELISPOT test is currently commercially available [24]. This test
utilizes both bovine tuberculin and ESTAT6/CFP10 for the identification of cats
infected with either M. bovis or M. microti and is able to differentiate the two mycobacteria. It is reported as having a sensitivity of 90% for detecting feline M. bovis
infections, 83.3% sensitivity for detecting feline M. microti infections, and 100%
specificity for both.
Serum antibody tests (multi-antigen print immune-assay (MAPIA), TB STAT-­
PAK, and Rapid DPP VetTB) have been evaluated in cats with TB [25]. Overall
sensitivity was 90% for detection of M. bovis infection and greater than 40% for M.
microti, with a specificity of 100%.
It is important to remember that these tests do not explicitly differentiate active
from latent infection or prior exposure. Culture of organisms from clinical samples
obtained from cats with appropriate signs and diagnostic findings remains the gold
standard for the diagnosis of active TB.
Treatment
Controlled studies of feline mycobacteriosis treatment are lacking, and the existing
literature consists of a few retrospective observational case series and case reports.
Mycobacterial Diseases
261
VetBooks.ir
Table 1 Drugs typically chosen to treat feline mycobacterial infections
Drug
Clofazimine
Dose
25 mg/cat PO q
24 h or 50 mg/
cat q 48 h
Clarithromycin
62.5 mg/cat PO
q 12 h
5–15 mg/kg PO
q 24 h
10 mg/kg PO q
24 h
Azithromycin
Rifampicin
Doxycycline
Enrofloxacin
Marbofloxacin
Orbifloxacin
Pradofloxacin
Moxifloxacin
5–10 mg/kg PO
q 12 h
5 mg/kg PO q
24 h
2 mg/kg PO q
24 h
7.5 mg/kg PO q
24 h
7.5 mg/kg PO q
24 h
10 mg/kg PO q
24 h
Side effects/comments
Skin and body fluid discoloration (pink-brown),
photosensitization, pitting corneal lesions, nausea,
vomiting, and abdominal pain
Possible hepatotoxicity
Monitor serum hepatic enzymesa
Cutaneous erythema and edema, hepatotoxicity, diarrhea,
and/or vomiting, neutropenia, thrombocytopenia
Vomiting, diarrhea, abdominal pain, hepatotoxicity
Hepatotoxicity and/or inappetence, cutaneous erythema/
pruritus, anaphylaxis
Monitor serum hepatic enzymesa
Hydrochloride or hyclate formulations may cause
esophageal irritation and possibly stricture
Enrofloxacin may cause retinal toxicity in cats;
marbofloxacin or orbifloxacin are preferred if available
Most M. avium complex organisms are resistant to
second-generation fluoroquinolones
Give without food unless gastrointestinal side effects
occur
Vomiting and anorexia; dose can be divided 12 hourly
and/or administered with food
Alanine transferase and alkaline phosphatase
a
There have been occasional reports of spontaneous resolution of M. lepraemurium
infection; [26, 27] however, the vast majority of SGM infections require treatment
to achieve a cure. Table 1 lists the drugs and doses typically chosen to treat feline
mycobacteriosis.
The initiation of empirical treatment is required in almost all cases of SGM
infection, as identification of the causative mycobacterium may take weeks to
months (or may not be available at all). The choice of initial treatment will depend
on [1] the suspected etiological agent, [2] owner factors such as finances and ability/
willingness to medicate the cat orally for an extended period, and [3] the presence
of comorbidities that may restrict the use of certain drugs, for example, hepatic
disease when using rifampicin.
Therapy should include at least rifampicin, clarithromycin (or azithromycin),
and/or pradofloxacin (or moxifloxacin). In areas where infection with the fastidious
organisms is common the inclusion of clofazimine, if available, would also be a
reasonable choice. Ethambutol and isoniazid have been used to treat feline TB,
although toxicity tends to limit their use. They tend only to be prescribed if there is
drug resistance to the more commonly utilized agents. If the infection is restricted
to a localized cutaneous site, surgical excision may be a beneficial adjunct to antibiotic therapy.
Medical treatment can be subsequently modified depending on identification of
the mycobacterial species involved, response to treatment, and/or, if available, the
VetBooks.ir
262
C. O’Brien
results of drug susceptibility testing. Therapy should extend for at least 2 months
post-surgical resection or beyond resolution of clinical signs. Unless diagnosed with
MTB complex infection, quarantine of the cat is not necessary. Some of the drugs,
especially clofazimine, induce photosensitivity; it is recommended that owners
keep the cat indoors in the summer months.
Prognosis
Localized cutaneous infection caused by all slowly growing species has a good
prognosis if treated promptly with a combination of appropriate antibiotics, and if
possible surgical resection. If cutaneous disease progresses to systemic infection,
the prognosis becomes significantly worse. Treatment is potentially expensive and
time-consuming. Cats can be notoriously tricky to medicate, and the provision of
multidrug therapy for many months may also affect the outcome.
Public Health Risks
The only SGM that appears to carry a definite risk of cat-to-human transfer is M.
bovis, although this risk seems to be low. A report from the UK details the infection
in four people (two clinically and two sub-clinically affected) associated with an
infected pet cat [28]. A laboratory worker seroconverted after exposure to research
cats that were infected after accidentally being fed infected meat [29]. At this time,
instances of cat-to-human transmission of M. microti infection have not been
reported.
There is one report of a person contracting M. marinum secondary to a cat scratch
[30]. However, this likely represented mechanical inoculation, rather than true zoonotic transfer. Likewise, there appears to be almost no risk of humans acquiring
infections from any of the fastidious organisms from cats; however, as the ecology
and transmission of these mycobacterial species are not understood, it is difficult to
determine their potential for zoonotic transfer completely.
The Advisory Board on Cat Diseases (based in Europe) recommends that all
people in contact with an infected cat should be made aware of the potential but low
risk of zoonotic transfer of feline mycobacteriosis [31]. As a minimal precaution,
the use of gloves is recommended when treating these animals. This is especially
important for anyone in contact with the cat who is immunocompromised. Veterinary
staff should utilize personal protective equipment when handling cats with cutaneous lesions, collecting biopsies, or performing necropsy studies.
References
1. Gupta RS, Lo B, Son J. Phylogenomics and comparative genomic studies robustly support
division of the genus Mycobacterium into an emended genus Mycobacterium and four novel
genera. Front Microbiol. 2018;9:67.
VetBooks.ir
Mycobacterial Diseases
263
2. Fedrizzi T, Meehan CJ, Grottola A, Giacobazzi E, Serpini GF, Tagliazucchi S, et al. Genomic
characterization of nontuberculous mycobacteria. Sci Rep. 2017;7:45258.
3. Lewis DT, Hodgin EC, Foil S, Cox HU, Roy AF, Lewis DD. Experimental reproduction of
feline Mycobacterium fortuitum panniculitis. Vet Dermatol. 1994;5(4):189–95.
4. Drancourt M, Raoult D. Cost-effectiveness of blood agar for isolation of mycobacteria. PLoS
Negl Trop Dis. 2007;1(2):e83.
5. Govendir M, Hansen T, Kimble B, Norris JM, Baral RM, Wigney DI, et al. Susceptibility of
rapidly growing mycobacteria isolated from cats and dogs, to ciprofloxacin, enrofloxacin and
moxifloxacin. Vet Microbiol. 2011;147(1–2):113–8.
6. Nash KA, Andini N, Zhang Y, Brown-Elliott BA, Wallace RJ. Intrinsic macrolide resistance in
rapidly growing mycobacteria. Antimicrob Agents Chemother. 2006;50(10):3476–8.
7. Brown-Elliott B, Philley J. Rapidly growing mycobacteria. Microbiol Spectr.
2017;5:TNMI7-0027-2016.
8. Malik R, Wigney DI, Dawson D, Martin P, Hunt GB, Love DN. Infection of the subcutis and
skin of cats with rapidly growing mycobacteria: a review of microbiological and clinical findings. J Feline Med Surg. 2000;2(1):35–48.
9. Guille AE, Tseng LW, Orsher RJ. Use of vacuum-assisted closure for management of a large
skin wound in a cat. J Am Vet Med Assoc. 2007;230(11):1669–73.
10. Vishkautsan P, Reagan KL, Keel MK, Sykes JE. Mycobacterial panniculitis caused by
Mycobacterium thermoresistibile in a cat. JFMS Open Rep. 2016;2(2):2055116916672786.
11. Ngan N, Morris A, de Chalain T. Mycobacterium fortuitum infection caused by a cat bite. N Z
Med J. 2005;118(1211):U1354.
12. Alves DM, da Motta SP, Zamboni R, Marcolongo-Pereira C, Bonel J, Raffi MB, et al.
Tuberculosis in domestic cats (Felis catus) in southern Rio Grande do Sul. Pesquisa Veterinária
Brasileira. 2017;37(7):725–8.
13. Gunn-Moore DA, McFarland SE, Brewer JI, Crawshaw TR, Clifton-Hadley RS, Kovalik M,
et al. Mycobacterial disease in cats in Great Britain: I. Culture results, geographical distribution and clinical presentation of 339 cases. J Feline Med Surg. 2011;13(12):934–44.
14. Cavanagh R, Begon M, Bennett M, Ergon T, Graham IM, De Haas PE, et al. Mycobacterium
microti infection (vole tuberculosis) in wild rodent populations. J Clin Microbiol.
2002;40(9):3281–5.
15. Delahay RJ, Smith GC, Barlow AM, Walker N, Harris A, Clifton-Hadley RS, et al. Bovine tuberculosis infection in wild mammals in the south-west region of England: a survey of prevalence
and a semi-quantitative assessment of the relative risks to cattle. Vet J. 2007;173(2):287–301.
16. Murray A, Dineen A, Kelly P, McGoey K, Madigan G, NiGhallchoir E, et al. Nosocomial
spread of mycobacterium bovis in domestic cats. J Feline Med Surg. 2015;17(2):173–80.
17. Fyfe JA, McCowan C, O'Brien CR, Globan M, Birch C, Revill P, et al. Molecular characterization of a novel fastidious mycobacterium causing lepromatous lesions of the skin, subcutis,
cornea, and conjunctiva of cats living in Victoria, Australia. J Clin Microbiol. 2008;46(2):
618–26.
18. O’Brien CR, Malik R, Globan M, Reppas G, McCowan C, Fyfe JA. Feline leprosy due to
Candidatus ‘Mycobacterium tarwinense’ further clinical and molecular characterisation
of 15 previously reported cases and an additional 27 cases. J Feline Med Surg. 2017;19(5):
498–512.
19. O'Brien CR, Malik R, Globan M, Reppas G, McCowan C, Fyfe JA. Feline leprosy due to
Candidatus ‘Mycobacterium lepraefelis’: further clinical and molecular characterisation of eight previously reported cases and an additional 30 cases. J Feline Med Surg.
2017;19(9):919–32.
20. Appleyard GD, Clark EG. Histologic and genotypic characterization of a novel Mycobacterium
species found in three cats. J Clin Microbiol. 2002;40(7):2425–30.
21. Miller MA, Fales WH, McCracken WS, O'Bryan MA, Jarnagin JJ, Payeur JB. Inflammatory
pseudotumor in a cat with cutaneous mycobacteriosis. Vet Pathol. 1999;36(2):161–3.
22. Malik R, Hughes MS, James G, Martin P, Wigney DI, Canfield PJ, et al. Feline leprosy: two
different clinical syndromes. J Feline Med Surg. 2002;4(1):43–59.
VetBooks.ir
264
C. O’Brien
23. Reppas G, Fyfe J, Foster S, Smits B, Martin P, Jardine J, et al. Detection and identification
of mycobacteria in fixed stained smears and formalin-fixed paraffin-embedded tissues using
PCR. J Small Anim Pract. 2013;54(12):638–46.
24. Rhodes SG, Gruffydd-Jones T, Gunn-Moore D, Jahans K. Adaptation of IFN-gamma ELISA and
ELISPOT tests for feline tuberculosis. Vet Immunol Immunopathol. 2008;124(3–4):379–84.
25. Rhodes SG, Gunn-Mooore D, Boschiroli ML, Schiller I, Esfandiari J, Greenwald R, et al.
Comparative study of IFNgamma and antibody tests for feline tuberculosis. Vet Immunol
Immunopathol. 2011;144(1–2):129–34.
26. O'Brien CR, Malik R, Globan M, Reppas G, Fyfe JA. Feline leprosy due to Mycobacterium
lepraemurium: further clinical and molecular characterization of 23 previously reported cases
and an additional 42 cases. J Feline Med Surg. 2017;19(7):737–46.
27. Roccabianca P, Caniatti M, Scanziani E, Penati V. Feline leprosy: spontaneous remission in a
cat. J Am Anim Hosp Assoc. 1996;32(3):189–93.
28. England PH. Cases of TB in domestic cats and cat-to-human transmission: risk
to public very low. 2014. Available from: https://www.gov.uk/government/news/
cases-of-tb-in-domestic-cats-and-cat-to-human-transmission-risk-to-public-very-low.
29. Isaac J, Whitehead J, Adams JW, Barton MD, Coloe P. An outbreak of Mycobacterium bovis
infection in cats in an animal house. Aust Vet J. 1983;60(8):243–5.
30. Phan TA, Relic J. Sporotrichoid Mycobacterium marinum infection of the face following a cat
scratch. Australas J Dermatol. 2010;51(1):45–8.
31. Lloret A, Hartmann K, Pennisi MG, Gruffydd-Jones T, Addie D, Belak S, et al.
Mycobacterioses in cats: ABCD guidelines on prevention and management. J Feline Med
Surg. 2013;15(7):591–7.
VetBooks.ir
Dermatophytosis
Karen A. Moriello
Abstract
Feline dermatophytosis is a superficial fungal skin disease of cats. The primary
mode of transmission is via direct contact or traumatic fomite inoculation.
Microsporum canis is the primary pathogen of cats although outdoor cats may
contract Trichophyton spp. infections. Diagnosis is based upon use of complementary diagnostic tests. Evidence-based studies have concluded there is no
one “gold standard diagnostic test.” Contrary to popular belief, evidence-based
studies found that Wood’s lamp examinations are positive in >91% of untreated
cats, making it a highly useful point-of-care diagnostic test when combined
with direct examination of hair and scales. PCR analysis of infective material
is also diagnostic. Fungal culture is needed for species identification. Topical
antifungal therapy is necessary to disinfect hairs, minimize disease transmission, and prevent environmental contamination. Systemic antifungal therapy
eradicates the disease within the hair follicle. Evidence-based studies have
shown that environmental disinfection is easily done via continued removal of
cat hair and debris. Spores do not multiply in the environment or invade homes;
spores are easily removed from soft and hard surfaces via washing with a detergent. Over-the-counter home disinfectants (i.e., bathroom cleaners) labelled as
efficacious against Trichophyton spp. are recommended over household bleach
which can be a human and animal health hazard. This is a low-level zoonotic
skin disease that may cause superficial skin lesions that are treatable and curable in people.
K. A. Moriello (*)
School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
e-mail: Karen.moriello@wisc.edu
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_13
265
266
K. A. Moriello
VetBooks.ir
Introduction
Dermatophytosis is a contagious, superficial fungal skin disease of skin, hair, scales,
and claws. It is non-life threatening, treatable, and curable and is a low-level zoonotic disease, i.e., it does not cause death and is easily treated. The disease will
resolve without treatment in otherwise healthy animals. Treatment is recommended
to shorten the course of the infection and limit the risk of transmission to other susceptible hosts. The two major goals of this chapter are to (1) summarize key aspects
of this disease from recent evidence-based studies and (2) provide evidence to counter many “Internet” myths surrounding this disease that result in poor treatment,
unwarranted client worries, and, in worst-case scenarios, euthanasia of cats and
kittens.
Pathogens of Importance and New Classifications
Dermatophytes are aerobic fungi that invade and infect keratinized skin, hair, scales,
and nails. These organisms are classified by host preference: anthropophilic
(humans), zoophilic (animals), and geophilic (soil).
Dermatophytes are also classified by different names depending upon whether or
not they are in an asexual state (anamorph) or a sexual state (teleomorph) [1, 2]. For
example, Microsporum canis is an anamorphic species that belongs to the teleomorphic Arthroderma otae complex (M. canis, M. ferrugineum, M audouinii) [3]. The
naming of anamorphs is based upon fungal culture macro- and micro-characteristics. Recently molecular testing has found many species to be one and the same. In
2011, the Amsterdam Declaration on Fungal Nomenclature (One Fungus = One
Name) was adopted, and reclassification is currently underway [4]. Trichophyton
and Microsporum are being reclassified into the genus Arthroderma. Clinicians
need to be aware of this, as clinical manuscripts are increasingly using new nomenclature. This chapter will use the traditional names.
The most important pathogen of cats is Microsporum canis. Less commonly cats
can be infected by Trichophyton spp. and M. gypseum. It has been well established
that dermatophytes are not part of the normal fungal flora of cats by both traditional
and molecular tests [5–7].
Prevalence
True disease prevalence is unknown as this is not a reportable disease. A recent
review of 73 papers from 29 countries revealed prevalence data was highly biased
depending upon the source of the cats, whether studies were prospective or retrospective, whether data was collected and interpreted before or after the recognition
of fomite carriage, and other inclusion criteria [2].
The most helpful data on prevalence is from studies where true disease was confirmed. These studies consistently found an overall low prevalence (<3%) in clinical
VetBooks.ir
Dermatophytosis
267
practice and shelters (Box 1). In one study from the United States (n = 1407 cats),
the overall prevalence of confirmed disease was 2.4% [8], while in a Canadian study
(n = 111 cats), it was 3.6% [9]. In a study from the United Kingdom (n = 154 cats),
it was 1.3% [10]. More interestingly, in another study from the United Kingdom, the
medical records of 142, 576 cats found that it was not even listed as a common skin
disease, even though 10.4% of cats were presented for skin disease [11]. In a study
looking at the chronic pruritic cats (n = 502), only 2.1% of cats were diagnosed with
the disease [12]. Finally, in a retrospective study of cats admitted to an open admission animal shelter (n = 5644), disease prevalence was 1.6% over a consecutive
24-month period of time [13].
Box 1: Key Points on Disease Prevalence
• Dermatophytosis is an uncommon cause of skin lesions in cats (<3%).
• It is untrue that “it is ringworm until proven otherwise!”
• It is a common skin disease in kittens.
Risk Factors
Risk factors for dermatophytosis include warm, humid environments, young
age, and group housing (e.g., animal shelters or catteries) [14–21]. Anecdotal
reports of “old age” or “older cats” with underlying age-related diseases being
predisposed to dermatophytosis were not supported by evidence [2]. Seropositive
FeLV or FIV cats have not been shown to be at increased risk of infection [22].
The development of dermatophytosis in cats receiving immunosuppressive
treatment for pemphigus foliaceus was not reported in two large studies [23,
24]. Given the widespread use of feline cyclosporine, there is only one cat
reported to have developed disease while receiving this drug [25]. With respect
to breed predispositions, Persian cats are often listed as being “predisposed”;
however, this breed is over-represented in prevalence and treatment studies.
Subcutaneous dermatophyte infections, although rare, are almost exclusively
reported in long haired breeds.
ey Aspects of the Pathogenesis, Transmission, and Immune
K
Response to Infection
Pathogenesis of Infection
The infective form of dermatophytes is an arthrospore which is formed by fragmentation of fungal hyphae into smaller infective units. There are three stages of a dermatophyte infection [2]. First, arthroconidia adhere to corneocytes which can occur
within 2–6 hours of exposure [26–28]. Second, fungi start to germinate with germ
VetBooks.ir
268
K. A. Moriello
tubes emerging from the arthroconidia followed by penetration of the stratum corneum. Finally, there is invasion of keratinized structures; dermatophyte hyphae
invade and grow in multiple directions including the hair follicle unit. Hyphae can
start to form arthroconidia within 7 days. Obvious clinical lesions are usually seen
within seven to 21 days.
Transmission
Development of M. canis lesions has been studied in direct application models of
infection and in co-habitant natural exposure experiments and offers practical
insights on disease transmission [29–35]. In direct application models, it was
extremely difficult to establish infections unless there was a critical mass of infective spores (>104 spores per site). Successful infection required micro-trauma and
occlusion of the site. It was not possible to infect cats/kittens that were allowed to
remove the infective inoculum via grooming. Positive Wood’s lamp fluorescence
was present in 100% of experimentally infected cats and was noted as early as 5 to
7 days post infection. In co-habitant models, a highly social infected cat was added
to a group of healthy cats, and lesion development followed a clear pattern. Lesions
developed over time in all cats, starting with the most social cats. All lesions began
on the face and ears and then progressed. In studies where healthy cats were housed
in contaminated environments but did not experience any skin micro-trauma, cats
became culture positive but did not develop lesions. Cats became culture negative
after washing or simply being moved to a clean room and allowed to groom.
It is now well established that the primary mode of disease transmission is via
direct contact with another infected animal. Grooming is an important innate protective mechanism against disease transmission. Micro-trauma is an important prerequisite for establishment of a successful infection. Increased micro-trauma to the
skin from pruritus or self-trauma, humidity, and ectoparasites all contribute to conditions optimal for disease development. Transmission from contaminated fomites
is a risk factor if it induces micro-trauma (e.g., grooming tools) or if the cat is in a
contaminated environment and self-traumatizes itself (e.g., is pruritic from ectoparasites). Transmission from contaminated environments is not an efficient mode of
transmission in the absence of micro-trauma and moisture.
Immunity and Recovery from Infection
Cats develop both a cell-mediated and humoral immune response to dermatophyte
infections [35–37]. Intradermal and in vitro studies show that recovery from infection depends upon the development of a strong cell-mediated immune response.
Cell-mediated immunity is important for protection against reinfection. Studies
have shown that reinfection of infected but cured cats was possible, but required a
greater number of spores, more occlusion, or both. The subsequent infections were
milder and resolved much sooner.
Dermatophytosis
269
VetBooks.ir
Clinical Findings
There are no “pathognomonic” clinical signs of feline dermatophytosis. The clinical
signs of dermatophytosis reflect the pathogenesis of the disease: invasion of keratinized structures of the skin. Clinical signs are also impacted by the age and overall
physiological health. For example, kittens with limited infections are at risk for
developing more widespread lesions if they contract upper respiratory infections or
gastrointestinal disease.
Practical Approach to “Clinical Signs”
Dermatophytosis severity reflects the overall global health of a cat. From that
perspective, there are different clinical presentations of dermatophytosis: simple
infections, complicated infections, and culture-positive lesion-free cats. Simple
infections are any disease that occurs in an otherwise healthy cat. Lesion severity
tends to be limited, and these cats respond well to treatment. It is likely that many
kittens develop limited lesions of dermatophytosis that self-resolve and never get
diagnosed. Complicated infections are more difficult to treat because lesions
tend to be more severe and the cat/kitten has a concurrent medical disease, develops a concurrent disease shortly after diagnosis that explains the severity of the
skin disease, and/or has some other complicating factor making treatment challenging (e.g., bandage requirements that make topical therapy options limited.)
A “complicated infection” is any case where treatment is not straightforward.
Culture-positive lesion-free cats are either cats that are fomite carriers or cats
that have subtle lesions that were missed at the time of initial examination. When
lesion-free, culture-positive cats are identified, re-examine the cat with a Wood’s
lamp and look for lesions. Fomite carriage is easily identified by simply washing
the cat, moving it to a clean environment, and repeating a culture. True fomite
carriage cats will be culture negative; a single bath will not remove infective
spores from the hair coat with true infection.
Common Findings
Lesions tend to be asymmetrical. As mentioned above, observational studies on cohabitant infection models documented that lesions tend to start on the face, ears, and
muzzle and then progress to the paws and tail (Figs. 1, 2, and 3) [34, 38]. Lesions
can be focal or multifocal. Hair loss may be mild, and sometimes, the primary client
concern is excessive hair loss. Some cats have a history of vomiting hair balls or
constipation. Scaling is common and sometimes can be marked (Figs. 4 and 5). In
severe cases, there can be exudative paronychia. The inflammatory reaction can
vary from mild to marked, and diffuse erythema may be present. Follicular plugging
and hyperpigmentation are somewhat uncommon in cats but most likely to be seen
in cats with dermatophytosis. Microsporum canis can cause comedone-like lesions
VetBooks.ir
270
K. A. Moriello
Fig. 1 Facial lesions on a
young cat with
dermatophytosis.
(Courtesy of Dr. Rebecca
Rodgers)
Fig. 2 Alopecia and
scaling on the pinna of a
Persian cat with
dermatophytosis.
(Courtesy of Dr. Chiara
Noli)
in young cats. Pruritus is variable and can be intense and may mimic areas of eosinophilic pyotraumatic dermatitis.
Uncommon Presentations
Uncommon clinical presentations in cats include cases clinically identical to pemphigus foliaceus, including symmetrical crusting over the face and ear and exudative paronychia. Unilateral or bilateral pinnal pruritus is another unique clinical
presentation. Infected hairs are on the ear margins or in the “bell” of the ear. Rarely,
VetBooks.ir
Dermatophytosis
Fig. 3 Alopecia on the
back and tail in an
advanced case of
dermatophytosis in a
Persian cat. (Courtesy of
Dr. Chiara Noli)
Fig. 4 Same cat as in
Figure 2: patch of alopecia
with very mild scaling.
(Courtesy of Dr. Chiara
Noli)
Fig. 5 Shorthaired cat
with dermatophytosis
showing a focal patch of
alopecia and thick scales.
(Courtesy of Dr. Chiara
Noli)
271
VetBooks.ir
272
K. A. Moriello
Fig. 6 Ulcerated nodule
due to a dermatophytic
mycetoma. (Courtesy of
Dr. Andrea Peano)
diffuse multifocal areas of waxy hyperpigmentation have been observed. Nodular
dermatophytosis has been most commonly reported in Persian cats (Fig. 6). There
is usually a history of prior dermatophytosis but not always. These may or may not
ulcerate and drain.
Diagnosis
Dermatophytosis cannot be diagnosed based upon clinical signs. A recent evidencebased review concluded that no one test could be identified as the “gold standard.”
Current recommendations are to use multiple complementary diagnostics [2].
Diagnostics for dermatophytosis are divided into two major categories: point-ofcare (POC) and reference laboratory (RL) testing. Complete blood counts, serum
chemistry panels, urinalysis, and diagnostic imaging are not helpful for confirming
the presence or absence of dermatophytosis. These tests are helpful when evaluating
a cat with a complicated infection.
Point-of-Care Diagnostics
There are three key complementary POC diagnostic tests and tools: dermoscopy,
Wood’s lamp examination, and direct examination of hair/scales. Dermoscopy and
Wood’s lamp examinations are tools used to find suspect hairs for direct examination. If infection can be confirmed via direct examination of hair shafts and scale,
treatment can be initiated at the time of presentation.
VetBooks.ir
Dermatophytosis
273
Dermoscopy
Dermoscopy (Figs. 7 and 8) is a non-invasive POC tool that allows for magnification and illumination of the skin. The primary use of dermoscopy is to find hairs
for direct examination. This tool can be used with or without a Wood’s lamp examination. Two studies found that M. canis-infected hairs have a unique appearance
[39, 40]. Infected hairs are opaque, slightly curved, or broken with a homogenous
thickness (Fig. 9). Hairs are easier to find in light colored cats than in darkly colored
cats. The biggest obstacle to the use of this test is patient cooperation.
ood’s Lamp Examination
W
A Wood’s lamp is a POC diagnostic tool whose primary usage is to find hairs for
direct examination or to lesion resolution in M. canis infected cats. A Wood’s lamp
is a plug-in lamp with an ultraviolet light spectrum of 320–400 nm wavelength [41]. In
Fig. 7 Hand-held
dermoscope. (Courtesy of
Dr. Fabia Scarampella)
Fig. 8 Cat being
examined with a
dermoscope. (Courtesy of
Dr. Fabia Scarampella)
VetBooks.ir
274
K. A. Moriello
Fig. 9 Infected hairs
visualized with a
dermoscope. (Courtesy of
Dr. Fabia Scarampella).
Arrows required to indicate
infected hairs
veterinary dermatology, the only fungal pathogen of importance that fluoresces is M.
canis. The characteristic green fluorescence of M. canis-infected hairs is due to a watersoluble pigment located within the cortex or medulla of the hair [42–44]. The fluorescence is the result of a chemical interaction that occurs as a result of the infection and
is not associated with spores or infective material.
An evidence-based review of literature has found that many commonly held
beliefs are incorrect about the usefulness of Wood’s lamp examination, prevalence
of positive fluorescence, and overall usefulness as a point-of-care “test”; this is tool
and it cannot be emphaized enough. Statements such as “less than 50% of strains
fluoresce” are based upon retrospective studies of random source diagnostic specimens [15, 45–47]. When data from 30 experimental infection studies and spontaneous disease studies was examined, results were surprisingly different [2]. There was
100% fluorescence in cats with experimental infections, and in studies involving
spontaneous disease in untreated animals, it was >91%. Not unexpectedly, positive
fluorescence was less common in cats under treatment. Fluorescing “tips” are a
common finding in cats that have been treated and cured. It is simply residual pigment left over from the infection within the hair follicle (Fig. 10).
In the author’s experience, the use of a Wood’s lamp is not unlike mastering
sample acquisition for skin/ear cytology and using a microscope. See Box 2 for
helpful hints on using a Wood’s lamp. It cannot be stressed enough that this is a skill
that can be learned. With the “right” Wood’s lamp and practice, this is a helpful tool
for finding suspect hairs (Figs. 11a, b). It is immensely helpful in finding lesions that
are otherwise missed in room light (Fig. 12). Fluorescing hairs are commonly found
in untreated infections; fluorescence may be more difficult to find in treated animals. False-positive and false-negative results are commonly due to inadequate
equipment, lack of magnification, patient compliance, poor technique, or lack of
275
VetBooks.ir
Dermatophytosis
SKIN
Fig. 10 Schematic of Wood’s lamp positive hairs (from left to right). Positive fluorescence is
found on the hair shafts only. Uninfected hairs show no fluorescence. Early infected hairs show
fluorescence in the proximal part of the hair. As the infection progresses, the entire hair shaft will
fluoresce. When the infection has been eradicated in the hair follicle, the proximal portion of the
hair shaft will no longer fluoresce (see hair with blue outline). This is an indication of a good
response to treatment. Cured cats will often have some residual “glowing tips” because the pigment is retained in the medulla or cortex (yellow = uninfected; green = fluorescent, thus infected).
Glowing tip hairs may or may not be culture positive
a
b
Fig. 11 Kitten with dermatophytosis: (a) mild lesions evident around the eyes; (b) evident fluorescence on the same sites with Wood light examination. (Courtesy of Dr. Laura Mullen)
VetBooks.ir
276
K. A. Moriello
Fig. 12 Wood’s lamp
positive hairs in the
interdigital space. This
lesion was not noticed
during examination in
room light
training. Wood’s lamp can rapidly identify high risk cats. For example, in one shelter, 1226 cats were surrendered in a 7-month period [48]. Of these cats, 273 (22.3%)
were culture positive, but only 60 of 273 were lesional, Wood’s lamp positive, and
direct examination positive. The 213 remaining culture-positive cats were nonlesional and Wood’s lamp negative and were determined to be fomite carriers. The
use of the Wood’s lamp at intake allowed for rapid identification of infected cats (50
of 60 being kittens).
Box 2: Wood’s Lamp Practice Tips
•
•
•
•
•
•
•
•
•
•
•
Use a medical grade lamp with UV spectrum 320 to 400 nm wavelength
Do not use hand-held battery-operated lamps
Use a lamp with built-in magnification
Lamps do not need to “warm up”
Allow your eyes to light adapt to the dark
Use a positive control slide
Hold lamp close to skin (2–4 cm); minimizes false fluorescence
Start at head and move slowly examining hair shafts
Lift crusts and look for apple green fluorescing hair shafts
Newly infected hairs are very short
Worried about false fluorescence? Examine hair bulb
irect Examination of Scales and Hairs
D
Direct examination is a POC diagnostic test that can confirm the presence of a dermatophyte infection at the time of initial presentation. Material can be collected
with the aid of a dermoscope and Wood’s lamp or via a combination of skin scraping and plucking of hairs. A recent study showed that the best way to collect
VetBooks.ir
Dermatophytosis
277
Fig. 13 Direct
examination of infected
hair (original magnification
4×). Note that infected
hairs are pale and
wider than normal hairs
which appear as 'threads' in
comparison.
specimens is via both superficial skin scraping and plucking of hairs from lesions.
In cats, combined hair plucking and skin scraping of lesion margins confirmed the
diagnosis in 87.5% of cases [49]. In this study, a Wood’s lamp was not used, and had
it been, the results may have been higher. The authors used mineral oil for mounting
of specimens and no clearing agent; clearing agents destroy fluorescence, cause
artifacts, and damage microscope lens. The author routinely mounts specimens in
mineral oil. This is a time and cost effective test to master as it allows for microscopic examination for mites with the same specimen. The most helpful aid in learning this technique is to have images comparing normal and abnormal hairs (Figs. 13,
14, and 15). Direct examination tips are summarized in Box 3.
Box 3: Direct Examination Practice Tips
Use a Wood’s lamp or dermoscope to help identify suspect hairs
Collect samples by both plucking and scraping of the lesion
Use a skin scraping spatula to scrape margins of lesions
Mount specimens in mineral oil; do not use a clearing agent
Use glass coverslips
Abnormal hairs are easily visible at 4× and 10×
Infected shafts are wider, paler, and often retractile compared to normal
hairs
Tips to find hairs
• Use a picture guide that shows abnormal vs normal hairs
• Hold a Wood’s lamp (2–3 cm) over the slide to help find the hairs
• Add lactophenol cotton blue or new methylene blue to the mineral oil; let
sample set for 10 to 15 minutes before examination; infected hairs will be
blue tinged
VetBooks.ir
278
K. A. Moriello
Fig. 14 Direct
examination of infected
(short thick arrow) and
uninfected hairs (long thin
arrow)
Fig. 15 Direct
examination of infected
hair in mineral oil and
lactophenol cotton blue
after 15 minutes
Skin Cytology
Macroconidia are never seen on cytological examination of skin cytology. However,
M. canis arthrospores may be observed in cats with severe infections (Fig. 16).
Fungal Culture
Fungal cultures can be a POC or RL diagnostic test (see Box 5). If infection is confirmed via direct examination of hair and scale, fungal culture is used to confirm the
dermatophyte species. Fungal culture can be used to confirm the diagnosis if POC
diagnostics do not. A recent study revealed good correlation between point-of-care
VetBooks.ir
Dermatophytosis
279
Fig. 16 Cytologic
examination of a skin
cytology from a cat with
dermatophytosis.
Numerous arthroconidia
are seen on the surface of
the corneocyte
cultures and reference laboratories when both gross colony formation along with
microscopic features were used to identify colonies [50]. However, there was an
almost 20% error rate when color change alone was used.
The most commonly used POC fungal culture medium is Dermatophyte Test
Medium (DTM) which consists of a nutrient medium plus inhibitors of bacterial
and saprophytic growth and phenol red as a pH indicator. Several variants are available, some of which claim to speed growth of the culture, but a study found that all
appeared to perform similarly [51]. From a practical perspective, use a POC plate
with a large volume of medium; it is easy to inoculate with a toothbrush or hairs and
is easily sampled for microscopic examination. The author discourages the use of
DTM glass vials because they are hard to inoculate and sample. Vials that base their
diagnostic value on “positive color change” are not recommended. Plates should be
stored in individual plastic bags to prevent cross contamination and protect against
desiccation and media mite infestation. The author stores samples in a plastic container and monitors temperature using an inexpensive digital thermometer for fish
tanks. Dermatophyte colonies may appear as soon as 5 to 7 days after inoculation.
Plates should be inspected daily for growth by holding the plate up to the light to
look for colony growth (Fig. 17). To minimize contaminant growth, do not open
plates until there is adequate growth to sample. It is common to see early colony
growth using this backlighting technique several days before a red color change
develops around the colony. The red color change is caused by a change in the pH
of the medium and is not diagnostic: it merely identifies colonies for microscopic
sampling (Fig. 17). The color change usually occurs at the time the colony is first
visible, but may develop within 12 to 24 hours after visible fungal growth. All fungal growth, including non-pathogens, will eventually produce a red media color
change after the colony has grown for several days to a week. Dermatophyte colonies are never green, gray, brown, or black. Pathogens are pale or buff in color and
have a powdery to cottony mycelial growth. All suspect colonies should be examined microscopically (Figs. 18 and 19).
VetBooks.ir
280
Fig. 17 DTM plate with
initial growth of M. canis.
Around the small white
cottony colonies, the
culture media color turned
red. Note: Gloves should
be worn when handling
fungal culture plates.
(Courtesy of Dr. Chiara
Noli)
Fig. 18 Microscopic
example of M. canis from
a clear acetate tape
preparation. The sample
was allowed to stand for
15 min before examination,
making macroconidia more
visible. Note the tapered
ends, rough surface, and
thick walls
Fig. 19 High power
(100×) of M. canis
macroconidia. Note the
thick walls as the most
consistent feature
K. A. Moriello
VetBooks.ir
Dermatophytosis
281
Unless there is a sampling error such as plucking only a few hairs rather than
using a toothbrush for sampling lesions, large numbers of colonies of the dermatophyte will appear on the plate if the animal is truly infected. The number of
colonies decreases as the infection resolves spontaneously or from treatment.
One of the most common problems with in-house culturing is the lack of sporulation or growth, or both, of M. canis on DTM. A common cause of this is overinoculation of the fungal culture plates. This is characterized by rapid swarming
of the plate with fluffy colony growth, but only unsporulated hyphae are noted on
microscopic examination. This can be avoided by limiting the number of toothbrush inoculations on the plate to six to eight stabs; individual impressions
should be clearly visible.
A common question is how long to hold fungal culture plates. Recent research
has shown that in cultures of human dermatophyte infections, 98.5% of fungal cultures were positive before day 17 [52]. A retrospective study of 2876 M. canis positive fungal cultures found that 98.2% were confirmed within 14 days of incubation
[53]. The revised commendation is to consider a fungal culture negative if no pathogen or no growth has been isolated by day 14.
Reference Laboratory Diagnostics
PCR
There is increasing interest and use of PCR in animals to diagnose dermatophytosis.
This is because PCR is commonly used to diagnose dermatophytosis of nail in people because of the difficulty in isolation of pathogens via fungal culture. Routine
bathing and use of over-the-counter topical antifungal preparations make isolation
of human Trichophyton infections via fungal culture challenging. If the reference
laboratory has the protocol, PCR on tissue can be used to aid in the diagnosis of
deep dermatophyte infections in cats [54, 55].
Commercial reference laboratories are increasingly offering PCR as a diagnostic test. The major advantage of this test over fungal culture is the rapid turnaround time. It is important to remember that PCR is very sensitive and will
detect both viable and non-viable fungal DNA. In addition, like toothbrush fungal cultures, PCR cannot differentiate between fomite carriage and true disease.
Field studies using a commercial PCR test in cats from animal shelters found that
the test has high sensitivity and high specificity [56, 57]. Samples can be collected using the toothbrush technique, but it is important to sample only the target lesion ensuring an adequate amount of follicular hair is collected for analysis.
Alternatively, avulsed crusts with hair shafts and hair bulbs can be collected and
submitted for examination. In one field study, the qPCR assay for Microsporum
spp. was more useful for initial disease confirmation, while the qPCR M. canis
assay was more useful for determining mycological cure [57]. When using this
test to monitor for mycological cure, it may be helpful to bathe and dry the cat’s
hair coat prior to sampling to minimize a positive PCR test from detection of
non-viable DNA.
VetBooks.ir
282
K. A. Moriello
Histopathology
There are two clinical presentations when histological examination of tissue is helpful to diagnose dermatophytosis. The first is when cats present with unusual skin
lesions and routine point-of-care diagnostics do not identify a cause. Some cats with
dermatophytosis will develop lesions that look clinically similar to pemphigus foliaceus. The second is the investigation of non-healing wounds or nodules caused by
dermatophytosis (dermatophytic mycetoma or pseudomycetoma). It is important to
remember to submit as large a section of tissue as possible because processing can
result in marked shrinkage of the specimen [58]. The author routinely samples nodules via excisional biopsy or with a 6 to 8 mm skin biopsy punch. It is important to
tell the pathology laboratory that dermatophytosis is suspected because routine
stains (i.e., hematoxylin and eosin) are not as sensitive as periodic acid-Schiff (PAS)
or Gomori’s methenamine silver (GMS) for detecting fungal elements in tissue. In
addition, tissue (4–6-mm punch or wedge) should be submitted to a reference laboratory for fungal culture.
Treatment
Dermatophytosis is a self-limiting disease in otherwise healthy animals. Treatment
is recommended to shorten the course of the infection because it is infectious and
contagious. Treatment is summarized in Table 1.
Confinement Considerations
Confinement needs to be reconsidered in the treatment of this disease. The recent
treatment consensus guidelines state that “Confinement needs to be used with care
and for the shortest time possible. Dermatophytosis is a curable disease, but behavior problems and socialization problems can be life-long if the young or newly
adopted animals are not socialized properly.” [2] This disease is most common in
kittens at the same time that it is critical for socialization and bonding. Veterinarians
need to consider animal welfare and quality of life when making a recommendation
for confinement. The purpose of confinement is to limit the amount of work needed
to do routine cleaning. The living area should allow for 24/7 exercise, normal behavior (i.e., play and jumping), sleeping, eating, and socialization. It is important to
remember that disease transmission is limited via the use of concurrent systemic
antifungal therapy and, most importantly, topical therapy. Infection from contaminated environments is an inefficient and rare mode of transmission. The infection is
transmitted by direct contact with spores on the hair coat. Topical therapy and simple barrier protection (i.e., gloves and long shirt sleeves) and reasonable human
behavior (i.e., not “wearing the kitten/cat”) will minimize the risk of transmission to
people.
Dermatophytosis
283
VetBooks.ir
Table 1 Quick summary of treatment recommendations
Confinement
Limit confinement to easily cleaned room. Kitten/cat must have 24/7 access to freely move
and exercise
Human interaction and socialization must be possible
Topical therapy
Twice weekly whole body therapy with lime sulfur or enilconazole rinses or shampoo therapy
with a miconazole-ketoconazole-climbazole/chlorhexidine shampoo
Daily focal miconazole 2% vaginal cream for lesions on the face
Daily otic medicaments that do not contain antibiotics for lesions in/on ears
Systemic therapy
Oral itraconazole 5 mg/kg orally once daily on a week on/week off basis: Do not use
compounded or reformulated itraconazole
Terbinafine 30 to 40 mg/kg orally once daily is an option if itraconazole is not available
Do not use lufenuron, griseofulvin, or fluconazole
Cleaning
“Clean as if company is coming twice weekly”
Keep cat hair to a minimum and use disinfectant wipes between “cleanings”
Hard surfaces
Focus on removal of cat hair and debris, wash with a detergent until visibly clean, rinse and
remove excess water, and use ready-to-use disinfectants labelled as effective against
Trichophyton spp. (i.e., bathroom cleaners)
Bleach is no longer recommended
Soft surfaces
Wash laundry twice to disinfect; do not over-stuff the washer tub and use the longest cycle
available
Vacuum carpets to remove cat hair; disinfect with steam cleaning or washing twice with a
beater brush carpet shampooer
Monitoring
Start monitoring after a cat/kitten is lesion-free and there are no fluorescing hair shafts
One fungal culture is compatible with mycological cure in most cats, two may be needed in
complicated situations, PCR can be used but may be associated with false positives
Clipping of the Hair Coat
There are no controlled studies comparing the number of days to cure in cats that
have been clipped to those that have not been clipped. Based upon treatment outcomes in dedicated dermatophyte treatment programs in the United States, it is the
author’s experience that routine clipping of the hair coat is not necessary. Clipping
of the hair coat requires sedation and can result in thermal burns which may not be
clinically apparent until weeks later. Based upon experimental treatment studies,
clipping of the hair coat can temporarily worsen lesions and/or result in the development of satellite lesions [30, 31]. If lesions or hair mats need to be removed, use
children’s round tipped metal scissors. Place the cat on newspaper to allow for easy
disposal of infective material. If long haired cats are slow to cure and/or the owner
VetBooks.ir
284
K. A. Moriello
cannot thoroughly soak the hair coat, scissor clipping to facilitate the penetration of
topical antifungal treatment may be helpful. What is helpful is to brush the hair coat
prior to the application of topical therapy to remove broken and easily shed hairs.
Plastic flea combs are ideal for this purpose.
Topical Therapy
Topical therapy is as important as systemic antifungal therapy in the treatment
of feline dermatophytosis. Systemic antifungal therapy eradicates infection
within the hair follicle but does not kill infective spores on or in the hair shaft or
on the hair coat. Topical therapy protects against disease transmission. Topical
therapy minimizes shedding of infective spores into the environment which
greatly decreases, if not prevents, the potential of positive fungal cultures due to
fomite contamination [59, 60]. Concurrent topical therapy will decrease the
overall length of treatment. It is important to remember that until the infection is
eradicated within the hair follicle, the hair coat will be continually reseeded with
infective spores.
Twice weekly whole body rinses or shampoos are recommended for the duration of treatment. Exposed uninfected cats and dogs should be treated with a
whole body antifungal rinse or shampoo to minimize risk of transmission. The
most consistently effective antifungal products in vivo studies are lime sulfur,
enilconazole, or miconazole/chlorhexidine shampoos [60–66]. These products
are fungicidal. The author has also successfully used combination miconazole/
chlorhexidine and climbazole/chlorhexidine leave-on mousse formulations in
cats that could not be wetted (i.e., cats with bandages, upper respiratory infections). In vitro studies have shown that antifungal shampoos containing miconazole, ketoconazole, climbazole, or accelerated hydrogen peroxide are antifungal
when used with a minimum of a 3-minute contact time [67]. There is strong
evidence from in vitro and in vivo studies on the antifungal activities of essential
oil preparations as options [59, 68, 69].
Based upon treatment of cats in dermatophyte treatment centers, the most
common reason for failure to cure is the presence of infective hairs in hard to
treat areas. People are reluctant to apply antifungal rinses or shampoo to the
face and in/near the ears of cats. Unfortunately, these are the sites of infective
hairs when “failure to cure” cats are examined with a Wood’s lamp. The author
recommends the daily use of miconazole 2% vaginal cream on the face and
periocular area if lesions are found. This particular product is used to treat fungal keratitis and is safe [70]. For infective hairs in the ears, antifungal otic products (chlorhexidine/miconazole or chlorhexidine/ketoconazole, or clotrimazole)
can be used daily.
Dermatophytosis
285
VetBooks.ir
Systemic Antifungal Therapy
Systemic antifungal therapy eradicates the infection within the hair follicle and is
used with concurrent topical therapy. Unless there is a contraindication, it is indicated in all cats with dermatophytosis.
The antifungal drug of choice for cats is itraconazole (Itrafungol, Elanco Animal
Health). It is labelled for use at 5 mg/kg orally once daily on an alternating week on/
week off treatment schedule. An initial treatment schedule of 3 cycles is recommended, but additional cycles may be needed in some cats that have not reached
mycological cure by 6 weeks [71]. Itraconazole accumulates in adipose tissue,
sebaceous glands, and hair for weeks after administration, making it suitable for
pulse therapy protocols [72].
Itraconazole has no age or weight limitations. Kittens as young as 10 days of age
were treated with 5 mg/kg orally for four consecutive weeks, and no treatmentrelated side effects were reported [72]. The drug is well tolerated, and no treatment
studies have reported death or adverse effects that required discontinuation of the
drug when used at doses for treatment of feline dermatophytosis [2]. Side effects
were rare and included salivation, mild anorexia, and vomiting [2, 71] Deaths associated with its use have not been reported. Target animal safety studies in which cats
were given 5, 15, and 25 mg/kg itraconazole for 7 days on alternate days for
17 weeks with an 8-week recovery period noted dose-related hypersalivation, vomiting, and loose stool which were mild to moderate and self-resolving [73].
Elevations in hepatic enzymes above baseline were sporadic, dose-related, and
rarely above laboratory normal ranges. In an extensive review of the literature,
reports of severe adverse effects of itraconazole in cats were all traced to studies or
case series treating cats with high doses for long periods of time [2].
The author is aware of many anecdotal reports of “itraconazole resistance,” and,
when investigated, compounded itraconazole was used. A recent paper compared
the reference capsule, reference solution, compounded capsule, and compounded
suspension in a randomized cross-over study [74]. The findings revealed that compounded formulations were poorly and inconsistently absorbed. Compounded itraconazole should not be used.
Terbinafine has been used successfully for the treatment of M. canis dermatophytosis [2]. Studies have shown that terbinafine is highly concentrated in the hair
coat of cats after 14 days of continuous administration and is suitable for pulse
therapy [75]. Doses in the literature range from 5 to 40 mg/kg per day; however,
higher doses of 30–40 mg have been reported to be clinically more effective. The
most common side effects are vomiting, diarrhea, and soft stools.
Griseofulvin was the first oral antifungal used to treat feline dermatophytosis, but it
is no longer recommended given the superior efficacy of itraconazole and terbinafine.
It is also a known teratogen and can cause dose-unrelated idiosyncratic bone marrow
VetBooks.ir
286
K. A. Moriello
suppression. Ketoconazole is effective against dermatophytes but is poorly tolerated in
cats and should not be used. Fluconazole has poor efficacy against dermatophytes and
should not be used. Numerous well-controlled studies have shown that lufenuron has
no efficacy and should not be used to treat dermatophytosis [32, 33, 76].
Fungal Vaccines
Antifungal vaccines for M. canis have shown no efficacy against challenge exposure but may be useful as adjuvant therapy. Commercial vaccines are limited in
availability [2].
Disinfection of the Environment
The primary reason to disinfect the environment is to minimize fomite contamination of the hair coat which will make it difficult to determine mycological cure.
Fomite contamination can lead to over-treatment of cats, over-confinement, expense,
and in some cases euthanasia. Review of the literature found that contact with a
contaminated environment alone in the absence of concurrent micro-trauma is a
rare source of infection for people and cats [2]. Severely contaminated environments, e.g., hoarding situations, are a risk factor for cats under severe physiological
stress or predisposed to skin micro-trauma (e.g., flea infestation).
Evidence-based studies on environmental decontamination have now shown that
it is much easier to decontaminate an environment than past literature suggests and/
or what clients may find on Internet resources. Dermatophyte spores can only live
and reproduce in keratin; they do not multiply or invade the environment as many
clients believe. It is important to stress to clients that dermatophyte spores are not
like mildew or mold that overgrows in homes after water damage. Clients will report
reading that “ringworm lives” in the environment up to 24 months. This comment
stems from a laboratory study where specimens (n = 25 total) were stored and sampled at various time points. In that study, three of six specimens stored between 13
and 24 months were viable on fungal culture medium [77]. This study did not document that stored specimens were able to cause disease. In a different study, stored
specimens were only viable for 13 months, and it was not possible to induce infection in kittens [78]. In the author’s experience with stored specimens for 25 years,
isolates loose viability and become culture negative within months. In one experiment, 30% (45 of 150 specimens) were culture negative within 5 months, and all
were culture negative by 9 months. Finally, spores in infective hairs and scales are
very susceptible to moisture: 100 specimens were culture negative after being
exposed to high humidity for 3 days.
Box 4 summarizes environmental cleaning recommendations. The environmental cleaning focus needs to be on mechanical cleaning and removal of debris coupled with washing of the target surface until visibly clean. The surface needs to be
rinsed of detergent as this will inactivate many disinfectants. In addition, the surface
needs to be free of any excess water as this will dilute disinfectants. A recent study
VetBooks.ir
Dermatophytosis
287
showed that household bathroom cleaners labelled as efficacious against
Trichophyton spp. are effective against the naturally infective form of M. canis and
Trichophyton spp. [79] Clients should be strongly advised against the use of household bleach as a disinfectant due its lack of detergency, lack of penetration into
organic material, and human/animal health hazard.
Box 4: Summary of Disinfection Recommendations
Key points to stress: Spores do not multiply in the environment, spores do not
invade surfaces like mildew or black mold, spores are easily removed via
cleaning, and spores are susceptible to moisture, i.e., they die quickly post
exposure.
Key cleaning points: “If you can wash it, you can decontaminate it” and
“clean as if company is coming.”
Cleaning specifics:
• Laundry: Wash twice in the washer on hot or cold water; bleach is not
necessary.
• Rugs: Keep pets off rugs and/or vacuum daily. Can be disinfected using
“steam cleaning” or washing twice with a beater brush carpet scrubber.
• Keep pets in easily cleaned rooms, but do not over-confine. Close closets
and drawers, and remove knick-knacks. Remove debris and pet hair daily
using dusting cloths or 3 M Easy Trap dust cloth (these are sticky “swiffers”)
and then mop floors with a flat mop. Repeat two to three times weekly.
• Disinfectants do not take the place of mechanical cleaning and washing;
spores are like dust and are easily removed via mechanical cleaning.
• Mechanical cleaning is most important, remove debris, wash with a detergent cleaner, rinse, and remove excess water. This alone can decontaminate
surface.
• Disinfectants are needed for spores not removed by cleaning. For safety,
only use ready-to-use commercial disinfectants labelled as efficacious
against Trichophyton spp., thoroughly wet target non-porous surfaces, and
let dry.
• Clean transport cages.
• Environmental sampling is NOT cost-effective and not recommended
unless there is concern about fomite contamination.
Clients often ask what they can do to minimize environmental contamination in
addition to cleaning. In addition to systemic antifungal therapy, the most important
thing is to use topical antifungal therapy to disinfect the hair coat. In a recent study,
proper cleaning combined with topical therapy resulted in homes being free of infective material within 1 week of starting treatment and remaining so throughout the
study [69]. In a study of 70 homes contaminated by M. canis infected cats, only 3 of
69 homes needed more than one cleaning for complete decontamination. One home
was never decontaminated due to admitted owner non-compliance [80].
288
K. A. Moriello
VetBooks.ir
Monitoring and Endpoint of Treatment
Mycological Cure
The term “mycological cure” was introduced into to the veterinary literature in
1959 and was defined as two negative fungal cultures at 2 weeks apart in a study
using griseofulvin to treat long haired cats with M. canis feline dermatophytosis
[81]. Because this is a contagious and infectious disease, M. canis is not part of the
normal fungal flora of cats, and the disease is of zoonotic importance, it is reasonable to treat cats until the infectious agent is no longer detectable via fungal
culture(preferred) or PCR. A recent study has found that the first negative fungal
culture was predictive of mycological cure in >90% of cats that were otherwise
healthy, where there was good compliance with cleaning, and topical and systemic
treatment [82].
Treatment Length
A common question from owners is “how long until the cat is cured”? The
response “as long as it takes” or “until mycological cure” is true, but irritating to
owners. In a recent placebo controlled study using itraconazole at current label
recommendations, lesion resolution, Wood’s lamp examinations, and weekly
fungal cultures were performed for 9 weeks [71]. In this study, cats did not
receive any topical therapy. Mycological cure was documented as early as week
four of treatment. At week nine, 39 of 40 (97.5%) cats had Wood’s negative
examinations. By the end of 9 weeks, 36 of 40 (90%) cats had at least 1 negative
fungal culture, and 24 of 40 had two negative fungal cultures. These cats did not
receive topical therapy. In a shelter study using 21 days of consecutive itraconazole and concurrent topical twice weekly lime sulfur, the mean number of days
to mycological cure was 18 (range 10–49 days) in a group of 90 cats that were
otherwise healthy [62]. In a later study involving random source cats with a multitude of concurrent illnesses, the mean number of days to cure was 37 days
(range 10–93) [83]. Based upon these studies, it is reasonable to answer the question as follows: in otherwise healthy cats receiving itraconazole and topical therapy, mycological cure can be expected within 4 to 8 weeks. If the cat has
concurrent illnesses, e.g., upper respiratory infection or poor nutrition, treatment
will be longer.
Recommended Monitoring
Clinical Cure
It is well established that clinical cure precedes mycological cure. There should be
resolution of clinical signs, and clients are usually capable of these observations. A
lack of resolution and/or development of new lesions indicate a treatment problem
or misdiagnosis. In the author’s experience, cats receiving itraconazole show rapid
resolution of clinical signs.
VetBooks.ir
Dermatophytosis
289
ood’s Lamp Examination
W
It is now known that Wood’s lamp examinations are very useful for both detection
of M. canis-infected hairs and for monitoring of infections. This tool is strongly
recommended to monitor infections provided the user has a proper Wood’s lamp,
cats can be handled, and the room darkened. As the infection is eradicated in the hair
follicle, fluorescence disappears from the proximal portion of the hair shaft (i.e.,
intra-follicular portion). As a new healthy hair grows, there is less and less fluorescence on the hair shaft. Residual pigment on the hair tips is common in cats that
have recovered from dermatophytosis and reflects residual pigment deposited in the
hair shaft at the time of initial infection.
PCR Testing
Commercial PCR testing for mycological cure can be used provided there is
high confidence in the reference laboratory performing the test. It is important
to remember that PCR will detect both viable and non-viable fungal
DNA. Leave-on rinses or mousses will kill fungal spores, but because these
are not “rinsed” off the hair coat, non-viable fungal DNA may be present.
Fungal PCR testing should be considered only after there is clinical cure and
lack of Wood’s lamp hair shaft fluorescence, routine cleaning is in place, and
only if topical therapy has been used concurrently with systemic antifungal
therapy. If a leave-on antifungal has been used for topical therapy, wash the
cat to remove any residual fungal DNA. qPCR M. canis assay was found to be
more useful for detecting mycological cure than the qPCR Microsporum assay
[57]. The use of cycle thresholds was found not to be helpful for determining
mycological cure [84].
Fungal Culture
The most commonly used diagnostic test to determine mycological cure in cats is a
toothbrush fungal culture. There is no established “best practice” for when to start
monitoring response to treatment using toothbrush fungal cultures. What is important to note is that it is no longer acceptable to report fungal culture results as “positive” or “negative.” The number of dermatophyte colony-forming units (cfu) per
plate provides valuable information (Boxes 5 and 6). Clinical examination, fungal
culture findings, and Wood’s lamp examination are used to determine if a cat is
infected or cured.
• Do weekly fungal cultures once the decision has been made to start evaluating
for mycological cure.
• Fungal culture plates do not need to be held longer than 14 days; culture negative
plates at day 14 should be considered negative.
• Do in-house cultures or use a reference laboratory that is familiar with the toothbrush inoculation technique and will provide weekly updates on cfu/plate.
• See Box 6 for use of cfu/plate and in practice.
• Continue topical therapy until the cat is mycologically cured (negative PCR or at
least one negative toothbrush fungal culture).
VetBooks.ir
290
K. A. Moriello
Box 5: Fungal Culture Practice Tips and Use of Colony-Forming Units
Fungal culture plates
• Use large volume easy open plates.
• Do not over-inoculate plates; make sure bristles show a pattern on
surface.
• Incubate in house in plastic bag to prevent cross contamination and minimize desiccation.
• Incubate at 25 °C to 30 °C.
• Examine daily for growth; use backlighting technique.
• No growth plates can be finalized at 14 days; not necessary to hold for
21 days.
Record growth twice weekly
• NG – no growth.
• C– contaminant growth bacterial or fungal.
• S – suspect growth (early growth of a pale colony or early growth of pale
colony with a red color change.
• Pathogen – requires microscopic identification. In animals under treatment, the red color change may lag behind the growth of the pale colony
especially as the animal approaches cure.
Count colony-forming units (only with toothbrush culture technique)
• The number of colony-forming units per plate can be used to monitor
response to therapy. P or “pathogen score” is the nickname used for this
system.
–– P3-≥ 10 cfu/plate (often too many to count!) – indicates high risk cat
and active infection
–– P2 5–9/cfu plate – indicates need to continue treatment
–– P1 1–4 cfu/plate – most consistent with fomite exposure or exposure to
another infected animal; continue topical therapy; improve cleaning of
environment; consider if there is exposure to infected animal
Note: This system makes it easy to monitor culture results and provides a
visual record of the pet’s response to treatment. In most cases, animals with
severe infections will have a starting culture score of P3. As treatment progresses, the P score becomes lower. Cured animals have cultures with no
growth or just contaminant on culture. The scoring system is also very helpful
in identifying pets undergoing treatment that are exposed to fomite contamination. These animals commonly will have cultures fluctuating from negative
to P1. When this pattern is seen, the owner can be instructed to improve
hygiene in the home. As fomite contamination is removed, the fungal cultures
become negative. In addition to identification of fomite exposure, this system
also rapidly alerts the clinician to animals that are failing therapy or are
VetBooks.ir
Dermatophytosis
291
relapsing for one reason or another. Lack of response to therapy will be suggested by a persistently high P score. Relapses will be represented by a sudden increase in colony-forming units.
Box 6: Interpretation of P-score, Lesions, and Wood’s Lamp Findings in
Diagnosis and Treatment of M. canis Infections∗
P-score
P3 (>10
cfu/
plate)
P2 (5–9
cfu/
plate)
P1 (1–4
cfu/
plate)
Wood’s lamp Wood’s
examination examination
Examination of hair shafts of hair tips Interpretation Plan
Lesional/
nonlesional
Positive/
negative
Positive/
negative
High risk/not
cured
Treat or
continue
treatment
Lesional
Positive/
negative
Positive/
negative
High risk/not
cured
Nonlesional
Positive
Positive/
negative
High risk/not
cured
Nonlesional
Negative
Positive/
negative
Cured/low
risk
Lesional
Positive/
negative
Positive/
negative
High risk/not
cured
Nonlesional
Positive
Positive/
negative
High risk/not
cured
Nonlesional
Negative
Positive/
negative
(glowing
tips are
common in
cured
animals)
Cured/low
risk
Treat or
continue
treatment
Treat or
continue
treatment
Re-examine,
apply whole
body
antifungal
treatment,
then repeat
culture when
dry
Treat or
continue
treatment
Treat or
continue
treatment
Re-examine,
apply whole
body
antifungal
treatment,
then repeat
culture when
dry
Comments
A single
infected hair
can produce a
P3 culture,
examine
carefully
Likely
represents a
“dust mop”
scenario
If “dust mop”
cat, repeat
culture will
be negative
Note
cfu colony forming unit; “dust mop” refers to a cat that is mechanically carrying spores from environmental contamination
∗
Adapted from the treatment and monitoring procedures used in the Felines In Treatment Program at the
Dane County Humane Society, Madison, Wisconsin, USA
Reprinted with permission from [2]
292
K. A. Moriello
VetBooks.ir
Public Health Aspects
Dermatophytosis was a disease of major public health concern because until relatively recently, there was no effective and safe antifungal treatment. Animalassociated infections were common because of people’s close association with
agriculture and the lack of veterinary care for skin diseases of pet animals. The
development of oral griseofulvin for use in people and small animals in the late
1950s was a major therapeutic advance for people and animals. The development of
ketoconazole, itraconazole, terbinafine and a wide range of topical antifungals were
further major advances.
Feline dermatophytosis is a pet-associated zoonosis, and it is a veterinarian’s
responsibility to inform clients of this risk and provide accurate information about
the disease. The reader is referred to the references for a detailed discussion [2]. Key
aspects to communicate to clients are the following:
• Dermatophytosis occurs in both animals and people. In people, it is commonly
called “toe nail fungus” or athlete’s foot fungus.
• It is the same disease, just a different pathogen. The primary pathogen of people
is Trichophyton.
• The disease causes skin lesions and is treatable and curable.
• From cats, the disease is transmitted via direct contact with hair or skin lesions,
and that is why topical therapy is so important. Topical therapy decreases the risk
of disease transmission.
• Use reasonable barrier protection, e.g., as you would in handling an animal with
infectious diarrhea.
• Risk of contracting the disease from the environment is low.
• Dermatophytosis is a common skin disease in immunocompromised people;
however, literature review found that these infections are resurgences of preexisting human dermatophyte infections [85]. Animal-associated dermatophytosis was rare.
• The most common complication of M. canis infection in immunocompromised
people was a prolonged treatment time [86].
References
1. Weitzman I, Summerbell RC. The dermatophytes. Clin Microbiol Rev. 1995;8:240–59.
2. Moriello KA, Coyner K, Paterson S, et al. Diagnosis and treatment of dermatophytosis in dogs
and cats.: Clinical Consensus Guidelines of the World Association for Veterinary Dermatology.
Vet Dermatol. 2017;28:266–e68.
3. Graser Y, Kuijpers AF, El Fari M, et al. Molecular and conventional taxonomy of the
Microsporum canis complex. Med Mycol. 2000;38:143–53.
4. Hawksworth DL, Crous PW, Redhead SA, et al. The Amsterdam declaration on fungal nomenclature. IMA Fungus. 2011;2:105–12.
5. Moriello KA, DeBoer DJ. Fungal flora of the coat of pet cats. Am J Vet Res. 1991;52:602–6.
VetBooks.ir
Dermatophytosis
293
6. Moriello KA, Deboer DJ. Fungal flora of the haircoat of cats with and without dermatophytosis. J Med Vet Mycol. 1991;29:285–92.
7. Meason-Smith C, Diesel A, Patterson AP, et al. Characterization of the cutaneous mycobiota
in healthy and allergic cats using next generation sequencing. Vet Dermatol. 2017;28:71–e17.
8. Scott DW, Miller WH, Erb HN. Feline dermatology at Cornell University: 1407 cases (1988–
2003). J Feline Med Surg. 2013;15:307–16.
9. Scott DW, Paradis M. A survey of canine and feline skin disorders seen in a university practice:
Small Animal Clinic, University of Montreal, Saint-Hyacinthe, Quebec (1987–1988). Can Vet
J. 1990;31:830.
10. Hill P, Lo A, Can Eden S, et al. Survey of the prevalence, diagnosis and treatment of dermatological conditions in small animal general practice. Vet Rec. 2006;158:533–9.
11. O’Neill D, Church D, McGreevy P, et al. Prevalence of disorders recorded in cats attending
primary-care veterinary practices in England. Vet J. 2014;202:286–91.
12. Hobi S, Linek M, Marignac G, et al. Clinical characteristics and causes of pruritus in
cats: a multicentre study on feline hypersensitivity-associated dermatoses. Vet Dermatol.
2011;22:406–13.
13. Moriello K. Feline dermatophytosis: aspects pertinent to disease management in single and
multiple cat situations. J Feline Med Surg. 2014;16:419–31.
14. Lewis DT, Foil CS, Hosgood G. Epidemiology and clinical features of dermatophytosis in
dogs and cats at Louisiana State University: 1981–1990. Vet Dermatol. 1991;2:53–8.
15. Cafarchia C, Romito D, Sasanelli M, et al. The epidemiology of canine and feline dermatophytoses in southern Italy. Mycoses. 2004;47:508–13.
16. Mancianti F, Nardoni S, Cecchi S, et al. Dermatophytes isolated from symptomatic dogs and
cats in Tuscany, Italy during a 15-year-period. Mycopathologia. 2002;156:13–8.
17. Debnath C, Mitra T, Kumar A, et al. Detection of dermatophytes in healthy companion dogs
and cats in eastern India. Iran J Vet Res. 2016;17:20.
18. Seker E, Dogan N. Isolation of dermatophytes from dogs and cats with suspected dermatophytosis in Western Turkey. Prev Vet Med. 2011;98:46–51.
19. Newbury S, Moriello K, Coyner K, et al. Management of endemic Microsporum canis dermatophytosis in an open admission shelter: a field study. J Feline Med Surg. 2015;17:342–7.
20. Polak K, Levy J, Crawford P, et al. Infectious diseases in large-scale cat hoarding investigations. Vet J. 2014;201:189–95.
21. Moriello KA, Kunkle G, DeBoer DJ. Isolation of dermatophytes from the haircoats of stray
cats from selected animal shelters in two different geographic regions in the United States. Vet
Dermatol. 1994;5:57–62.
22. Sierra P, Guillot J, Jacob H, et al. Fungal flora on cutaneous and mucosal surfaces of cats infected
with feline immunodeficiency virus or feline leukemia virus. Am J Vet Res. 2000;61:158–61.
23. Irwin KE, Beale KM, Fadok VA. Use of modified ciclosporin in the management of feline
pemphigus foliaceus: a retrospective analysis. Vet Dermatol. 2012;23:403–e76.
24. Preziosi DE, Goldschmidt MH, Greek JS, et al. Feline pemphigus foliaceus: a retrospective
analysis of 57 cases. Vet Dermatol. 2003;14:313–21.
25. Olivry T, Power H, Woo J, et al. Anti-isthmus autoimmunity in a novel feline acquired alopecia
resembling pseudopelade of humans. Vet Dermatol. 2000;11:261–70.
26. Zurita J, Hay RJ. Adherence of dermatophyte microconidia and arthroconidia to human keratinocytes in vitro. J Invest Dermatol. 1987;89:529–34.
27. Vermout S, Tabart J, Baldo A, et al. Pathogenesis of dermatophytosis. Mycopathologia.
2008;166:267–75.
28. Baldo A, Monod M, Mathy A, et al. Mechanisms of skin adherence and invasion by dermatophytes. Mycoses. 2012;55:218–23.
29. DeBoer DJ, Moriello KA. Development of an experimental model of Microsporum canis
infection in cats. Vet Microbiol. 1994;42:289–95.
30. DeBoer D, Moriello K. Inability of two topical treatments to influence the course of experimentally induced dermatophytosis in cats. J Am Vet Med Assoc. 1995;207:52–7.
VetBooks.ir
294
K. A. Moriello
31. Moriello KA, DeBoer DJ. Efficacy of griseofulvin and itraconazole in the treatment of experimentally induced dermatophytosis in cats. J Am Vet Med Assoc. 1995;207:439–44.
32. Moriello KA, Deboer DJ, Schenker R, et al. Efficacy of pre-treatment with lufenuron for the
prevention of Microsporum canis infection in a feline direct topical challenge model. Vet
Dermatol. 2004;15:357–62.
33. DeBoer DJ, Moriello KA, Blum JL, et al. Effects of lufenuron treatment in cats on the establishment and course of Microsporum canis infection following exposure to infected cats. J Am
Vet Med Assoc. 2003;222:1216–20.
34. DeBoer DJ, Moriello KA. Investigations of a killed dermatophyte cell-wall vaccine against
infection with Microsporum canis in cats. Res Vet Sci. 1995;59:110–3.
35. Sparkes AH, Gruffydd-Jones TJ, Stokes CR. Acquired immunity in experimental feline
Microsporum canis infection. Res Vet Sci. 1996;61:165–8.
36. DeBoer DJ, Moriello KA. Humoral and cellular immune responses to Microsporum canis in
naturally occurring feline dermatophytosis. J Med Vet Mycol. 1993;31:121–32.
37. Moriello KA, DeBoer DJ, Greek J, et al. The prevalence of immediate and delayed type hypersensitivity reactions to Microsporum canis antigens in cats. J Feline Med Surg. 2003;5:161–6.
38. Frymus T, Gruffydd-Jones T, Pennisi MG, et al. Dermatophytosis in cats: ABCD guidelines on
prevention and management. J Feline Med Surg. 2013;15:598–604.
39. Scarampella F, Zanna G, Peano A, et al. Dermoscopic features in 12 cats with dermatophytosis
and in 12 cats with self-induced alopecia due to other causes: an observational descriptive
study. Vet Dermatol. 2015;26:282–e63.
40. Dong C, Angus J, Scarampella F, et al. Evaluation of dermoscopy in the diagnosis of naturally
occurring dermatophytosis in cats. Vet Dermatol. 2016;27:275–e65.
41. Asawanonda P, Taylor CR. Wood’s light in dermatology. Int J Dermatol. 1999;38:801–7.
42. Wolf FT. Chemical nature of the fluorescent pigment produced in Microsporum-infected hair.
Nature. 1957;180:860–1.
43. Wolf FT, Jones EA, Nathan HA. Fluorescent pigment of Microsporum. Nature.
1958;182:475–6.
44. Foresman A, Blank F. The location of the fluorescent matter in microsporon infected hair.
Mycopathol Mycol Appl. 1967;31:314–8.
45. Sparkes A, Gruffydd-Jones T, Shaw S, et al. Epidemiological and diagnostic features of
canine and feline dermatophytosis in the United Kingdom from 1956 to 1991. Vet Rec.
1993;133:57–61.
46. Wright A. Ringworm in dogs and cats. J Small Anim Pract. 1989;30:242–9.
47. Kaplan W, Georg LK, Ajello L. Recent developments in animal ringworm and their public
health implications. Ann N Y Acad Sci. 1958;70:636–49.
48. Newbury S, Moriello K, Coyner K, et al. Management of endemic Microsporum canis dermatophytosis in an open admission shelter: a field study. J Feline Med Surg. 2015;17:342–7.
49. Colombo S, Cornegliani L, Beccati M, et al. Comparison of two sampling methods for
microscopic examination of hair shafts in feline and canine dermatophytosis. Vet (Cremona).
2010;24:27–33.
50. Kaufmann R, Blum SE, Elad D, et al. Comparison between point-of-care dermatophyte test
medium and mycology laboratory culture for diagnosis of dermatophytosis in dogs and cats.
Vet Dermatol. 2016;27:284–e68.
51. Moriello KA, Verbrugge MJ, Kesting RA. Effects of temperature variations and light exposure on the time to growth of dermatophytes using six different fungal culture media inoculated with laboratory strains and samples obtained from infected cats. J Feline Med Surg.
2010;12:988–90.
52. Rezusta A, Gilaberte Y, Vidal-García M, et al. Evaluation of incubation time for dermatophytes
cultures. Mycoses. 2016;59:416–8.
53. Stuntebeck R, Moriello KA, Verbrugge M. Evaluation of incubation time for Microsporum
canis dermatophyte cultures. J Feline Med Surg. 2018;20:997–1000.
54. Bernhardt A, von Bomhard W, Antweiler E, et al. Molecular identification of fungal pathogens
in nodular skin lesions of cats. Med Mycol. 2015;53:132–44.
VetBooks.ir
Dermatophytosis
295
55. Nardoni S, Franceschi A, Mancianti F. Identification of Microsporum canis from dermatophytic pseudomycetoma in paraffin-embedded veterinary specimens using a common PCR
protocol. Mycoses. 2007;50:215–7.
56. Jacobson LS, McIntyre L, Mykusz J. Comparison of real-time PCR with fungal culture for the
diagnosis of Microsporum canis dermatophytosis in shelter cats: a field study. J Feline Med
Surg. 2018;20:103–7.
57. Moriello KA, Leutenegger CM. Use of a commercial qPCR assay in 52 high risk shelter cats
for disease identification of dermatophytosis and mycological cure. Vet Dermatol. 2018;29:66.
58. Reimer SB, Séguin B, DeCock HE, et al. Evaluation of the effect of routine histologic processing on the size of skin samples obtained from dogs. Am J Vet Res. 2005;66:500–5.
59. Nardoni S, Giovanelli S, Pistelli L, et al. In vitro activity of twenty commercially available, plant-derived essential oils against selected dermatophyte species. Nat Prod Commun.
2015;10:1473–8.
60. Paterson S. Miconazole/chlorhexidine shampoo as an adjunct to systemic therapy in controlling dermatophytosis in cats. J Small Anim Pract. 1999;40:163–6.
61. Moriello K, Coyner K, Trimmer A, et al. Treatment of shelter cats with oral terbinafine and
concurrent lime sulphur rinses. Vet Dermatol. 2013;24:618–e150.
62. Newbury S, Moriello K, Verbrugge M, et al. Use of lime sulphur and itraconazole to treat
shelter cats naturally infected with Microsporum canis in an annex facility: an open field trial.
Vet Dermatol. 2007;18:324–31.
63. Carlotti DN, Guinot P, Meissonnier E, et al. Eradication of feline dermatophytosis in a shelter:
a field study. Vet Dermatol. 2010;21:259–66.
64. Jaham CD, Page N, Lambert A, et al. Enilconazole emulsion in the treatment of dermatophytosis in Persian cats: tolerance and suitability. In: Kwochka KW, Willemse T, Von Tscharner
C, editors. Advances in Veterinary Dermatology. Oxford: Butterworth Heinemann; 1998.
p. 299–307.
65. Hnilica KA, Medleau L. Evaluation of topically applied enilconazole for the treatment of dermatophytosis in a Persian cattery. Vet Dermatol. 2002;13:23–8.
66. Guillot J, Malandain E, Jankowski F, et al. Evaluation of the efficacy of oral lufenuron combined with topical enilconazole for the management of dermatophytosis in catteries. Vet Rec.
2002;150:714–8.
67. Moriello KA. In vitro efficacy of shampoos containing miconazole, ketoconazole, climbazole or accelerated hydrogen peroxide against Microsporum canis and Trichophyton species. J
Feline Med Surg. 2017;19:370–4.
68. Mugnaini L, Nardoni S, Pinto L, et al. In vitro and in vivo antifungal activity of some essential
oils against feline isolates of Microsporum canis. J Mycol Med. 2012;22:179–84.
69. Nardoni S, Costanzo AG, Mugnaini L, et al. Open-field study comparing an essential oil-based
shampoo with miconazole/chlorhexidine for haircoat disinfection in cats with spontaneous
microsporiasis. J Feline Med Surg. 2017;19:697–701.
70. Gyanfosu L, Koffuor GA, Kyei S, et al. Efficacy and safety of extemporaneously prepared miconazole eye drops in Candida albicans-induced keratomycosis. Int Ophthalmol. 2018;38:2089–210.
71. Puls C, Johnson A, Young K, et al. Efficacy of itraconazole oral solution using an alternating-­
week pulse therapy regimen for treatment of cats with experimental Microsporum canis infection. J Feline Med Surg. 2018;20:869–74.
72. Vlaminck K, Engelen M. An overview of pharmacokinetic and pharmacodynamic studies
in the development of itraconazole for feline Microsporum canis dermatophytosis. Adv Vet
Dermatol. 2005;5:130–6.
73. Elanco US I. Itrafungol itraconazole oral solution in cats. Freedom of Information Summary
NADA 141–474, November 2016.
74. Mawby DI, Whittemore JC, Fowler LE, et al. Comparison of absorption characteristics of oral
reference and compounded itraconazole formulations in healthy cats. J Am Vet Med Assoc.
2018;252:195–200.
75. Foust AL, Marsella R, Akucewich LH, et al. Evaluation of persistence of terbinafine in the hair
of normal cats after 14 days of daily therapy. Vet Dermatol. 2007;18:246–51.
VetBooks.ir
296
K. A. Moriello
76. DeBoer D, Moriello K, Volk L, et al. Lufenuron does not augment effectiveness of terbinafine for treatment of Microsporum canis infections in a feline model. Adv Vet Dermatol.
2005;5:123–9.
77. Sparkes AH, Werrett G, Stokes CR, et al. Microsporum canis: Inapparent carriage by cats and
the viability of arthrospores. J Small Anim Pract. 1994;35:397–401.
78. Keep JM. The viability of Microsporum canis on isolated cat hair. Aust Vet J. 1960;36:277–8.
79. Moriello KA, Kunder D, Hondzo H. Efficacy of eight commercial disinfectants against
Microsporum canis and Trichophyton spp. infective spores on an experimentally contaminated
textile surface. Vet Dermatol. 2013;24:621–e152.
80. Moriello KA. Decontamination of 70 foster family homes exposed to Microsporum canis infected
cats: a retrospective study. Vet Dermatol. 2019;30:178–e55. https://doi.org/10.1111/vde.12722.
81. Kaplan W, Ajello L. Oral treatment of spontaneous ringworm in cats with griseofulvin. J Amer
Vet Med Assoc. 1959;135:253–61.
82. Stuntebeck RL, Moriello KA. One vs two negative fungal cultures to confirm mycological cure
in shelter cats treated for Microsporum canis dermatophytosis: a retrospective study. J Feline
Med Surg. 2019. https://doi.org/10.1177/1098612X19858791.
83. Newbury S, Moriello KA, Kwochka KW, et al. Use of itraconazole and either lime sulphur or
Malaseb Concentrate Rinse (R) to treat shelter cats naturally infected with Microsporum canis:
an open field trial. Vet Dermatol. 2011; 22: 75–9.
84. Jacobson LS, McIntyre L, Mykusz J. Assessment of real-time PCR cycle threshold values
in Microsporum canis culture-positive and culture-negative cats in an animal shelter: a field
study. J Feline Med Surg. 2018;20:108–13.
85. Rouzaud C, Hay R, Chosidow O, et al. Severe dermatophytosis and acquired or innate immunodeficiency: a review. J Fungi. 2015;2:4.
86. Elad D. Immunocompromised patients and their pets: still best friends? Vet J. 2013;197:662–9.
VetBooks.ir
Deep Fungal Diseases
Julie D. Lemetayer and Jane E. Sykes
Abstract
Deep mycotic infections are uncommon in cats. However, in endemic regions,
cryptococcosis, sporotrichosis, and histoplasmosis occur regularly in immunocompetent cats. Cryptococcosis and sporotrichosis are more prevalent in cats
than in dogs, and histoplasmosis is as prevalent or possibly slightly more prevalent in cats than in dogs. Blastomycosis and coccidioidomycosis are rare in cats,
even in highly endemic areas. Sino-nasal and sino-orbital aspergillosis are also
infrequent worldwide but interestingly, brachycephalic cats appear to be predisposed. Lastly, infections with saprophytic opportunistic fungi usually result from
an accidental cutaneous inoculation in otherwise immune-competent cats and
cause localized signs. Occasionally, however, disseminated infections can occur.
Cats with systemic mycosis frequently have cutaneous manifestations. Reported
cutaneous signs include multifocal ulcerated or nonulcerated cutaneous masses,
subcutaneous masses, and draining abscesses, among others. The cutaneous
signs are frequently associated with systemic signs of illness and/or other organ
involvement, which should raise suspicion for fungal infection. The present
chapter focus on the epidemiology, clinical signs, including cutaneous signs,
diagnostic tests, and treatment of clinically important systemic fungal infections
in cats. In addition, it reviews the antifungal drugs currently available for the
treatment of these infections.
J. D. Lemetayer (*) · J. E. Sykes
Veterinary Medical Teaching Hospital, University of California, Davis, CA, USA
e-mail: jesykes@ucdavis.edu
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_14
297
298
J. D. Lemetayer and J. E. Sykes
VetBooks.ir
Introduction
Deep mycotic infections are uncommon to rare in cats worldwide. A 1996 study
estimated a prevalence of seven deep mycotic infections per 10,000 cats in the USA
[1]. Indeed, cats are relatively resistant to fungal infections and the prevalence of
most fungal infections is lower in cats than in dogs except for cryptococcosis and
sporotrichosis. Cats also may be slightly more susceptible to histoplasmosis than
dogs [2]. This chapter will focus on the epidemiology, clinical signs, diagnostic
tests for, and treatment of, clinically important systemic fungal infections in cats.
Box 1: Dimorphic Fungi
• Cryptococcosis, caused by C. gattii and C. neoformans, is the most common fungal disease in cats worldwide.
• Histoplasmosis is seen as frequently as or slightly more frequently in cats
than in dogs in endemic regions
• Cats are also more susceptible to sporotrichosis.
• Blastomycosis and coccidioidomycosis are both infrequent in cats.
• Most cats are immunocompetent.
• Cutaneous signs are frequent in these dimorphic fungal diseases.
• Fluconazole is the first line treatment for most cases of cryptococcosis, and
itraconazole is used for resistant cases of cryptococcosis (mostly C. gattii
infections) and infections with other dimorphic fungal organisms.
• A combination with amphotericin B is recommended in severe cases.
• A short course of anti-inflammatory dose of glucocorticoids is recommended for CNS cases and animals with severe pulmonary disease.
Cryptococcosis
Epidemiology
The most common fungal infection in cats is cryptococcosis [1]. Cryptococcus spp.
are dimorphic basidiomycetous fungi. Two main species cause cryptococcosis in
cats: Cryptococcus neoformans and Cryptococcus gattii. Rarely, other species have
been implicated. Cryptococcus magnus was isolated from a cat with otitis externa
in Japan [3] and in a cat with a deep limb infection in Germany [4]. Cryptococcus
albidus was isolated from a cat with disseminated cryptococcosis in Japan [5]. Two
of these three cats were tested for feline immunodeficiency virus (FIV) and feline
leukaemia virus (FeLV) and were negative [4, 5], and no other apparent underlying
immunocompromise was identified in the three cats.
Cryptococcus neoformans is the most common species of Cryptococcus isolated worldwide and include two varieties: C. neoformans var. neoformans and
VetBooks.ir
Deep Fungal Diseases
299
C. neoformans var. grubii. Cryptococcus neoformans var. grubii accounts for the
majority of cases in Australia [6].
Cryptococcus gattii is mostly found in the west coast of the United States and in
British Columbia, Canada; in South America, southeast Asia (New Guinea, Thailand),
and in parts of Africa and Australia. While C. neoformans is more common than C.
gattii in Australia, rural cats in Australia and cats from Western Australia seem to be
more commonly infected by C. gattii than by C. neoformans [6, 7].
Cryptococcus neoformans can be found in avian guano, especially pigeon faeces; but is also found in other sources including milk, fermenting fruit juices, air,
dust and decaying vegetation [6]. C. gattii is often found in the hollows of trees,
especially Eucalyptus trees, in Australia, but has been associated with other hardwood tree species in other geographic locations.
Cryptococcus spp. have been divided into molecular types. Cryptococcus neoformans var. grubii isolates belong to molecular types VNI and VNII, whereas C.
neoformans var. neoformans isolates belong to molecular type VNIV [7]. A hybrid
variety of serotype AD has been classified as molecular type VNIII. C. gattii isolates are classified as VGI, VGII, VGIII, and VGIV. There is a proposal to rename
Cryptococcus molecular types as separate species of Cryptococcus, which remains
controversial.
Clinical Features in Cats
Siamese, Birman, Ragdoll, Abyssinian, and Himalayan breeds seem to be overrepresented in studies evaluating cats with cryptococcosis [1, 6, 8–10], although
this was not found in a study from California [11]. A male predisposition has been
identified in a few studies [10, 12] but not in others [6, 9, 11]. Having access to the
outdoors is also likely a risk factor but cats kept strictly indoors can also be affected
[8]. Cats of all age are affected, and FIV or FeLV status does not seem to be a risk
factor [6].
The incubation period is variable and can range from months to many years in
animals which were initially able to control the disease [13]. After inhalation of the
fungus, many cats have upper respiratory involvement with chronic sneezing, nasal
discharge and nasal deformation and/or deformations of the structures adjacent to
the nasal cavities such as the sinuses (Fig. 1). Involvement of the nasal cavity has
been reported in 43–90% of cases [1, 6, 12]. Infections also involve the retina,
draining lymph nodes and the central nervous system (CNS). Clinical signs include
enlarged mandibular lymph nodes, blindness, dilated and fixed pupils, slow pupillary light reflexes, lethargy, ataxia, behavioural changes and disorientation. Single
or multifocal ulcerated or nonulcerated cutaneous masses were seen in 31% and
41% of cases in two studies [1, 12]. The masses can be firm or fluctuant, raised,
dome-shaped and erythematous. They frequently ulcerate and may ooze a greyish
gelatinous exudate [1]. Other cutaneous lesions include plaques, miliary papules,
firm, dome-shaped, alopecic and erythematous papules or nodules [14]. Cutaneous
lesions are usually an extension of a sino-nasal pathology. Involvement of the skin
VetBooks.ir
300
J. D. Lemetayer and J. E. Sykes
Fig. 1 Cat with nasal
cryptococcosis caused by
Cryptococcus gattii
and subcutis in multiple sites suggests dissemination of the infection. Other uncommon locations include the lungs (2–12% of cases), [1, 6, 15] gingiva [15], salivary
glands [6], middle ear [16], kidneys, periarticular subcutaneous tissues, footpads
and bones [6].
Diagnostic Tests
Changes on complete blood count (CBC), serum biochemistry and urinalysis are
mild and non-specific. [17] A specific diagnosis of cryptococcosis can be obtained
with antigen detection using latex agglutination assays on serum. The assay can
also be used on pleural or peritoneal effusions, urine, and cerebrospinal fluid
(CSF). The clinical sensitivity of the test on serum in cats ranges from 90% to
100% and specificity ranges from 97% to 100% [18]. The sensitivity appears to be
lower in dogs. If the antigen test is negative, and cryptococcosis is still a possibility, tissue samples should be submitted for cytology, histology and culture [11].
When titres are <1:200, confirmatory tests are strongly recommended. Enzymelinked immunosorbent assays (ELISAs) are also under study but data are not currently available.
VetBooks.ir
Deep Fungal Diseases
301
Fig. 2 India ink negative
stain highlighting the
Cryptococcus
polysaccharide capsule
On cytology, cryptococcal yeasts are encapsulated, spherical to oval yeasts measuring 4–10 μm with narrow-based budding. The thick mucopolysaccharide capsule
is a major virulence factor for the pathogen because it allows the organism to hide
from the host immune system. It appears as a clear halo in stained smears and can
be visualized using India ink negative stains (Fig. 2) [19]. However, the capsule
can vary in size and in some patients the capsule can be thin [20] which can sometimes complicate the diagnosis. In these cases, there can be morphological overlaps
between Histoplasma and Cryptococcus on cytology [20].
On histopathology, yeasts may be associated with well-ordered granulomas or
pyogranulomas, sometimes with a background of a few eosinophils, lymphocytes
and plasma cells. Lesions may also contain a large number of yeasts and only a
mild degree of inflammation. This results in a “soap bubble” appearance on haematoxylin and eosin staining (H&E) [14] due to the organism’s thick, non-staining,
capsule. Macroscopically, these lesions are gelatinous masses (cryptococcomas).
In skin biopsies, numerous organisms are often present in the dermis, panniculus
and subcutis [14] but occasionally, less typical lesions can complicate the diagnosis.
For example, in a case series of four cats with cutaneous cryptococcosis, severe
granulomatous to pyogranulomatous cutaneous lesions were reported with large
numbers of eosinophils, but organisms could not be seen using H&E stain in three
of the four cats, and the organisms were capsule deficient [14].
When yeasts are not seen using H&E stain, special stains such as Grocott’s
methenamine silver (GMS), periodic acid–Schiff (PAS), Fontana-Masson stain,
Ziehl–Neelsen stain or Mayer’s mucicarmine stains may help reveal organisms.
Polymerase chain reaction (PCR) assays can also be applied to fresh biopsies
or formalin-fixed paraffin-embedded tissues [14]. False-positive PCR results can
VetBooks.ir
302
J. D. Lemetayer and J. E. Sykes
occur from nasal tissues since subclinical nasal cavity colonization can occur, and
therefore positive results should always be considered with the rest of the clinical
picture [17].
Fresh tissue can also be submitted for fungal culture. Cryptococcus spp. grow on
most laboratory media in 2 to 10 days. Because the organisms grow in culture as a
yeast, rather than a mould, on routine fungal media they are less likely to represent
a laboratory hazard than organisms that grow as moulds [17].
Treatment and Prognosis
The triazoles are the first line therapy in the treatment of cryptococcosis and can be
used as a monotherapy for mild to moderate cryptococcal infections. Fluconazole is
often preferred to itraconazole because of its good penetration in the brain, eye, and
urinary tract; lower cost; and minimal adverse effects. However, the development
of fluconazole resistance has been reported during treatment [21, 22]. Resistance
to fluconazole can be due to overexpression or a change in the copy number of
ERG11, the gene encoding for the target enzyme 14-α-demethylase [21, 22]. Efflux
of triazole drugs via multidrug efflux transporters (AFR1 for C. neoformans and
PDR11 for C. gattii VGIII) has also been identified. Isolates that are resistant to
fluconazole remain susceptible to itraconazole but may demonstrate moderate voriconazole resistance [23].
In severe cases such as those with CNS involvement, addition of amphotericin B
to either fluconazole or itraconazole is recommended [24]. While the penetration of
amphotericin B into the CNS and vitreous is poor, the blood-brain or blood-eye barrier is compromised at the beginning of treatment so a clinical response can still occur.
Flucytosine can also be used in combination with amphotericin B because of the synergy between the two anti-fungal drugs and because of flucytosine’s good penetration
in the CNS, however it may be cost prohibitive. See Tables 1 and 2 for more information regarding anti-fungal drugs in cats. A short course of prednisolone may improve
outcome for cats with CNS infection because it decreases CNS inflammation at the
beginning of anti-fungal treatment and may help limit neurological deterioration [25].
Typically, at least 6 to 8 months of treatment is necessary, and often treatment
needs to be continued for years [17]. Serial monitoring of serum antigen titres
should be used to evaluate treatment response as a decline in titre correlates with
elimination of organisms [12]. Treatment should be continued until the titre is zero.
Unfortunately, relapse can still occur after successful treatment and after titres have
become negative, sometimes as long as 10 years after therapy is discontinued [24].
Prognosis is generally good, with the possible exception of cats with CNS infection
[24, 25]. The prognosis may also depend on the Cryptococcus species and molecular
type. For example, it is the authors’ experience that infections with C. gattii VGIII tend
to be less likely to be cured than those with VGII. While FeLV likely has a negative
impact on the treatment response, the effect of FIV status on outcome is not clear. Good
response to treatment is often seen despite a positive FIV status but these cats may have
more severe disease and/or may respond more slowly to treatment [9, 10, 26].
Decreased
antifungal
activity
Clinical use
Mechanism
of action
Dose
(continued)
Ketoconazole
Fluconazole
Itraconazole
Voriconazole
Posaconazole
Inhibition of 14α-demethylase, a CYT P450-dependant fungal enzyme = accumulation of 14α-methylsterols and disruption of the fungal
cell membrane
Oral suspension: 30 mg/kg
Not available
50 mg/cat PO q
25–50 mg/cat PO or IV 5 mg/kg PO q 12–24 h,
12–24 h
q 12–24 h
100 mg capsule q 48 h [129] Possible dose: 12.5 mg/cat once then 15 mg/kg q 48 h,
or 15 mg/kg once then
PO every 72 hours (with
or 3 mg/kg q 12–24 h with
extreme caution as may be 5–7.5 mg/kg q 24 h [90,
oral solution. Do not use
120, 132]
compounded solutions [130] associated with severe
toxicity) [131]
Yeasts, dimorphic fungi,
Yeasts, dimorphic fungi,
Dimorphic fungi,
Malassezia spp.,
Candida spp.,
most moulds, including
most moulds especially
Aspergillus spp. and some
Dimorphic fungi
Malassezia spp.,
zygomycosis
Aspergillus spp.
other moulds
Coccidioides spp.,
Cryptococcus spp.,
Histoplasma spp.
Sporothrix schenckii,
Poor activity against Aspergillus spp.
zygomycosis
intrinsically resistant,
many moulds,
poor activity against
including
many moulds
Aspergillus spp.
Histoplasma:
Development of
resistance during
treatment
Table 1 Azole antifungal drugs used in cats
VetBooks.ir
Deep Fungal Diseases
303
Strong inducer CYT
P450: many drug
interactions
Additional
comments
Itraconazole
Good distribution into skin,
bone & lungs. Limited
penetration to CNS, eyes
and kidneys/urine
Reported in 25% of cases
[134]. Gastrointestinal signs,
hepatotoxicity and lethargy
For the capsules: give with
food and avoid antacid
medications.
Therapeutic drug monitoring
at steady state (14-21d)
[135] recommended
Fluconazole
Widely distributed
including eyes, CNS
and kidneys/urine
[133]
Well tolerated.
Gastrointestinal signs,
hepatotoxicity
uncommon
Very good oral
absorption
CNS central nervous system, CYT P450 cytochrome P 450, PO per os, h hours, d days
Common:
Gastrointestinal
signs, hepatotoxicity
Ketoconazole
Good penetration
into most tissues but
not CNS
Adverse
effects
Tissue
distribution
Table 1 (continued)
Gastrointestinal signs and
increased liver enzyme
activities
Visual changes (miosis),
ataxia, paralysis,
hypersalivation,
hypokalaemia and
arrhythmias [131]
Give without food.
Therapeutic drug
monitoring recommended
(trough concentration).
Strong inducer CYT P450:
many drug interactions
Give with food and avoid
antacid medications. Low
oral absorption. Therapeutic
drug monitoring
recommended (trough
concentration)
Posaconazole
Widely distributed but
probably not in urine
Voriconazole
Widely distributed
including eyes, CNS and
kidneys/urine
VetBooks.ir
304
J. D. Lemetayer and J. E. Sykes
Decreased
antifungal
activity
Tissue
distribution
Clinical use
Doses
Mechanism
of action
Some Aspergillus spp. Poor
efficacy against Pythium
insidiosum
Poor penetration of the CNS
& eyes. Liposomal and lipid
complex formulations have
better CNS penetration and
less nephrotoxicity
Amphotericin B
Formation of pores in the
fungal cell membrane by
binding to sterols = leakage
of ions
Deoxycholate AmB: 0.25 mg/
kg IV or 0.5 mg/kg SC
AmB lipid complex and
liposomal AmB: 1 mg/kg IV
3 times weekly (up to 12
treatments)
Yeasts, dimorphic fungi and
most moulds
Reported resistance for some
dermatophytes [137] and
Aspergillus spp. [138]
Concentrate in skin nails and
hairs
Dermatophytes, maybe useful
in combination with other
antifungal drugs for various
mould infections
Terbinafine
Inhibition of squalene
epoxidase = reduction of
ergosterol production in the
fungal membrane
30–40 mg/kg PO q 24 h
Table 2 Other clinically important anti-fungal drugs in cats
(continued)
Never used as sole agent because
of rapid development of
resistance
Widely distributed including
eyes, CNS
Cryptococcus spp. and Candida
spp.
Invasive aspergillosis refractory to
other antifungal therapy, invasive
candidiasis. Some activity against
Histoplasma spp. and Coccidioides
spp. Variable activity against other
filamentous fungi
Cryptococcus spp., Fusarium spp.,
Rhizopus spp. and Mucor spp. are
resistant [139]
Widely distributed. Poor penetration
of the CNS & eyes
1 mg/kg IV once then 0.75 mg/kg q
24 h [136]
Flucytosine
Deamination of flucytosine to
5-fluorouracil = interfere with
DNA replication and protein
synthesis
25–50 mg/kg PO q 6–8 h
Caspofungin
Inhibition of β-1,3-D-­
glucans = disturb the integrity of the
fungal cell wall
VetBooks.ir
Deep Fungal Diseases
305
Amphotericin B
Cumulative nephrotoxicity
(mostly AmB deoxycholate),
rarely haemolytic anaemia
[140].
Sterile injection site abscesses
with SC injections
Liposomal and lipid complex
formulations have better CNS
penetration and less
nephrotoxicity
Low oral absorption [141]
Terbinafine
Well tolerated. Rarely, GI
toxicity and facial pruritus
Caspofungin
Possible anaphylactic reaction.
Transient fever and diarrhoea
reported [136]
AmB Amphotericin B, CNS central nervous system, GI gastro-intestinal, IV intravenous, PO per os, SC subcutaneous
Additional
comments
Adverse
effects
Table 2 (continued)
Avoid in animals with renal
failure
Flucytosine
Myelosuppression and GI signs
VetBooks.ir
306
J. D. Lemetayer and J. E. Sykes
Deep Fungal Diseases
307
VetBooks.ir
Histoplasmosis
Epidemiology
Histoplasma capsulatum is a dimorphic, soil-borne fungus that is endemic in the
USA (especially in the central and eastern states but has also been described in
California and Colorado), Central and South America, Africa, India and Southeast
Asia [27, 28]. It is found worldwide in various mammalian species but besides cases
in these endemic areas, cases of histoplasmosis in cats have only been described in
Ontario, Canada [29], Thailand [30] and Europe (Italy, Switzerland) [28, 31]. In a
1996 study, histoplasmosis was the second most common fungal disease in cats in
the USA with an incidence of 0.01% of the total feline hospital population of the
veterinary medical database [1].
Histoplasma capsulatum has been divided into eight to nine geographic clades by
multi-locus sequence typing: North American-1, with possibly a related phylogenetically distinct strain isolated from non-endemic American areas; North American-2;
Latin American group A; Latin American group B; Australian; Netherlands (of
Indonesian origin); Eurasian; and African [32].
The primary reservoir of H. capsulatum is the intestinal tracts and guano of bats.
It can also be found in decaying avian guano (especially around blackbird or starling roosts and chicken coops). After inhalation or ingestion, the fungus transforms
into a yeast phase within the body of cats and is engulfed by phagocytic cells, primarily macrophages. Trafficking of these cells results in dissemination of the yeasts
via the blood and lymphatics away from the lung and gastrointestinal tract to organs
of the mononuclear phagocyte system mostly (lymph nodes, liver, spleen, and bone
marrow) as well as other tissues. Yeast are 2–4 μm in diameter and are surrounded
by a 4 μm thick wall and reside within mononuclear phagocytes [33].
Clinical Features in Cats
Cats of all age can be affected with a mean age, of 4 and 9 years in two studies [1,
34]. Persian cats may be slightly over-represented [1]. A sex predisposition has not
been clearly identified, but females were over-represented in one case series [34].
Most cats are not concurrently infected with either FeLV or FIV. The disease seems
to be diagnosed more often between the months of January to April [1] and can also
affect cats that are housed exclusively indoors [35]. The reported duration of clinical signs before diagnosis of histoplasmosis ranged from 2 weeks to 3 months. [1]
When cats have clinical histoplasmosis, disseminated disease is the most commonly reported clinical presentation [36]. Clinical signs exhibited by cats with disseminated disease are mostly non-specific and include lethargy, weight loss, fever,
anaemia, dehydration, weakness and anorexia [1, 34]. Respiratory signs such as
dyspnoea and tachypnoea are common, but cough is rare. Other common clinical signs include hepatomegaly, icterus, lymphadenopathy and splenomegaly [36,
37], ocular signs (chorioretinitis, anterior uveitis, or retinal detachments) [1, 29,
VetBooks.ir
308
J. D. Lemetayer and J. E. Sykes
38] and skeletal involvement (lameness or swelling of one or more limbs) [1, 38,
39]. Clinical signs of gastrointestinal tract involvement such as vomiting, diarrhoea,
melaena or haematochezia are less common than in dogs [2]. Less common sites of
infection include the skin [28, 38, 40, 41], CNS [42], oral mucosa [43] and urinary
bladder [44].
Cutaneous signs consist usually of multiple papules and nodules which may be
ulcerated and exude serosanguineous fluid. A case of cutaneous fragility secondary
to disseminated histoplasmosis has also been described [41]. The cat had a large
skin tear that developed over the dorsal cervical region with epidermal atrophy,
dermal collagen separation, and infiltration in the dermis and subcutis with macrophages and intravascular monocytes containing Histoplasma yeasts on histology.
Diagnostic Tests
CBC findings include anaemia, which is often normocytic and normochromic and
non-regenerative [10, 34]. Thrombocytopenia is also reported, as well as leucocytosis and leukopenia. Occasionally, H. capsulatum may be seen within phagocytic
cells on peripheral blood smears from dogs and cats [1]. On serum biochemistry,
hypoalbuminemia is a common finding. Cats with liver involvement can have
increased liver enzyme activity and hyperbilirubinemia. Hyperglobulinemia and
azotaemia are also reported in few cats [33], as well as hypercalcaemia [45].
Abnormalities on thoracic radiographs are common and may be subclinical [1,
44]. Radiographic patterns in cats with pulmonary histoplasmosis include fine,
diffuse or linear interstitial patterns, bronchointerstitial patterns, diffuse miliary
or nodular interstitial patterns, alveolar patterns and/or areas of pulmonary consolidation [33]. Sternal lymphadenopathy is also reported [46]. Bone lesions on
radiographs are typically osteolytic, but there may be periosteal and endosteal proliferative lesions, which are mostly found in appendicular bones with a predilection
for the elbow and stifle joints [39].
A definitive diagnosis of histoplasmosis is made by cytologic or histopathologic
identification of H. capsulatum in tissues (Fig. 3). The organisms are usually identified intracellularly within macrophages but can sometimes be found free in necrotic
exudates and may be confused with Cryptococcus spp. [20] As for Cryptococcus
infections, a variety of stains can highlight the yeasts, such as Diff-Quik and Wright
stains for cytology and GMS or PAS stains for histology.
The yeasts can be found on cytology of lymph nodes, lung, liver, spleen, skin
or bone marrow. Serum antibody assays are available but their clinical utility has
been limited by low sensitivity and specificity [47]. An antigen ELISA assay has
been evaluated for the diagnosis and monitoring of histoplasmosis in cats when
applied to serum and urine specimens [46, 47]. Sensitivities of 93–94% of the assay
were reported in two studies when applied to urine whereas the sensitivity was
only 73% when applied to serum [46, 47]. A specificity of 100% was found in
one of these two studies, which included 20 cats diagnosed with other non-fungal
diseases [47]. Based on the human literature, serologic cross-reactivity with other
VetBooks.ir
Deep Fungal Diseases
309
Fig. 3 Cytology showing
intracellular Histoplasma
yeast organisms
fungal pathogens such as Blastomyces spp. is however expected [47]. Antigen concentrations decrease with effective anti-fungal treatment and increase in cases that
were not well controlled or following relapse [46]. However, antigen elimination
sometimes preceded clinical remission and four cats still had measurable antigen
concentrations at the time of remission.
Fungal culture and PCR can also be used to confirm a diagnosis of histoplasmosis. However, fungal culture is a hazard to laboratory workers and should therefore
be performed only if necessary, and the laboratory should be warned of the possibility of a dimorphic fungal infection, so that appropriate precautions are taken.
Although most cultures are positive within 2 or 3 weeks, growth may require up to
6 weeks of incubation. PCR is currently not used routinely for diagnosis but was
used in a few cases to confirm the diagnoses in non-endemic areas [27, 28, 30, 48].
It can also be used when the identity of the fungus observed on histopathology is
in doubt.
Treatment and Prognosis
Itraconazole is the treatment of choice for histoplasmosis [45]. Treatment is recommended for a minimum of 4 to 6 months and should be continued for at least
2 months after resolution of clinical signs and possibly until antigen assays are negative. The use of itraconazole may be cost-prohibitive for some clients and adverse
effects are more common than with fluconazole, particularly hepatotoxicity [35]. A
retrospective study comparing the outcome of 17 cats treated with fluconazole to
13 cats treated with itraconazole found no difference in mortality and relapse rate
between the two groups suggesting that fluconazole may be a suitable alternative
[35]. However, a lower efficacy of fluconazole compared to itraconazole, and development of fluconazole resistance during treatment has been described in people
VetBooks.ir
310
J. D. Lemetayer and J. E. Sykes
[49] and in a cat [50]. The fluconazole-resistant isolates also had increased MICs to
voriconazole but not to itraconazole or posaconazole.
Deoxycholate or lipid-complexed amphotericin B can be used initially to treat
cats with severe acute pulmonary, acute disseminated, or CNS disease, after which
treatment should be continued with either itraconazole or fluconazole. Other possible treatment options include posaconazole in cats that do not tolerate itraconazole
or those that fail to respond to fluconazole. A short course of anti-inflammatory glucocorticoids may be useful for cats with severe pulmonary disease or CNS disease
at the beginning of treatment.
The prognosis depends on the extent of disease, with reported survival rates
varying from 66% to 100% [35, 45].
Blastomycosis
Epidemiology
Blastomyces dermatitidis is also a dimorphic fungus. It is found as a mycelium in
the environment and as a thick-walled budding yeast in tissues [51]. Blastomycosis
is a rare disease in cats and most feline infections are identified at necropsy. A
1996 study identified 41 cases over a 30–year time frame, and blastomycosis constituted 0.005% of all feline cases in the veterinary medical database [1]. In North
America, cases of blastomycosis are mostly found in the eastern and southern parts
of the USA, especially the Ohio and Mississippi River Valleys, and in the Great
Lakes region, as well as in Canada, especially Quebec, Ontario, Manitoba, and
Saskatchewan [1, 52–54]. Blastomycosis is also endemic in Africa and India [51].
Blastomycosis was also reported in a cat from Thailand [55].
In endemic regions, B. dermatitidis is found in localized regions where soils
are moist and acidic with decaying vegetation or animal excreta [52]. Inhalation of
conidia produced from the mycelial phase in soil or decaying matter is the primary
route of infection [51]. Direct inoculation of the organism via skin puncture wounds
occurs rarely.
From the lungs, the organism may disseminate via the vascular or lymphatic
system, resulting in a granulomatous or pyogranulomatous inflammatory response
in many organs, especially the lymph nodes, eyes, skin, bones and brain.
Clinical Features in Cats
A male predisposition to blastomycosis was found in one study and cats less than
4 years of age appear predisposed [1, 53, 56]. However in another case series of
eight cats, most cases were female and over 7 years of age [52]. In addition, Siamese,
Abyssinian, and Havana Brown cats may be predisposed [1]. Immunosuppression
does not seem to play a role in predisposition to the disease [52], and cats housed
strictly indoors can also be affected [52, 57, 58].
VetBooks.ir
Deep Fungal Diseases
311
Fig. 4 Lateral thoracic
radiograph of a cat with
pulmonary blastomycosis.
Courtesy University of
California, Davis
Veterinary Medical
Teaching Hospital
Diagnostic Imaging
Service
Duration of illness at diagnosis ranges from less than 1 week to 7 months and
clinical signs include dyspnoea, cough, anorexia, lethargy, weight loss, peripheral
lymphadenopathy, lameness, cellulitis of the limbs, CNS and cutaneous and ocular
signs (Fig. 4) [1, 51]. Cutaneous involvement occurred in 23% of 22 affected cats
[1], in 63% of eight cats in another case series [52] and in six additional cats [57,
59]. Cutaneous signs include non-ulcerated dermal masses, ulcerative skin lesions,
draining abscesses or cellulitis [1, 52, 59].
Diagnostic Tests
Bloodwork findings in cats with blastomycosis are non-specific and indicative of an
inflammatory process, such as mild non-regenerative anaemia [52]. Hypercalcaemia
and increased calcitriol concentration have also been reported [59]. A specific diagnosis of blastomycosis is usually made using cytologic examination of impression
smears, lavage specimens, or aspirates (skin, lymph nodes or lungs). Cytology was
diagnostic in 4/6 cases and in 4/5 cases in two studies [53, 57]. Blastomyces yeasts
are round to oval, 10–20 μm in diameter, have a basophilic cytoplasm, thick and
double-contoured walls and display broad-based budding [51]. A pyogranulomatous inflammatory reaction is typical, but a suppurative response predominates on
occasion. Special stains such as PAS, Gridley’s fungal, and GMS can assist in the
detection of the yeasts. Histology of tissue or bone biopsies, fungal culture or PCR
can also be used to make a diagnosis of blastomycosis. However, culture is time-­
consuming and a hazard for laboratory staff. To date, PCR assays have primarily
been used for research purposes [60, 61] but in one report, PCR was used to confirm
a diagnosis of blastomycosis in a cat from a non-endemic country [55].
Serology has not been well evaluated in cats. In a study, only one of four cats
with blastomycosis tested positive on agar gel immunodiffusion (AGID) using
Blastomyces whole cell antigen [1].
312
J. D. Lemetayer and J. E. Sykes
VetBooks.ir
Treatment and Prognosis
Cats with blastomycosis are usually treated with itraconazole. Fluconazole appears
to be less effective than itraconazole, but it may be a more appropriate treatment
choice for urinary tract, prostatic and CNS infections because of improved penetration of these organs. The addition of amphotericin B for cats with severe disease
such as CNS or severe disseminated disease may also be valuable [56, 62]. However,
two cats with severe disease treated with amphotericin B did not respond well to
treatment [63]. Newer triazoles, including voriconazole, posaconazole and isavuconazole, have activity against B. dermatitidis. Voriconazole and posaconazole have both
been successfully used to treat severe blastomycosis in humans, especially cases with
CNS involvement [64], but in general voriconazole should be avoided in cats due to
their susceptibility to voriconazole toxicity. Clinical signs and radiographic lesions
can worsen in the first few days of treatment as a result of the inflammatory response
to dying organisms. A short course of glucocorticoids at an anti-inflammatory dose
should be considered for patients with CNS involvement and severe respiratory disease, although whether this ultimately improves outcome is not known.
In one study, 4 of 8 cats responded favourably to either itraconazole or fluconazole
[52] and 4 of 7 responded favourably to surgical resection of cutaneous lesions and
administration of ketoconazole and potassium iodide in another study [1]. However
only 1 of 4 cats survived with itraconazole treatment in another study [57].
Coccidioidomycosis
Epidemiology
Coccidioidomycosis is also a rare disease worldwide in cats. The largest case series
(48 cats) was reported from Arizona, including 41 cases that were diagnosed over
3 years [65]. Coccidioidomycosis is endemic to the semiarid desert regions of
the southwestern USA, southern and central California, southern Arizona, southern New Mexico, western Texas, southern Nevada and Utah, as well as northern
Mexico, and parts of Central and South America [66].
Coccidioidomycosis is caused by Coccidioides immitis and Coccidioides posadasii. Coccidioides immitis is mostly found in California and C. posadasii is
found elsewhere [67]. No significant differences in morphology or disease course
have been noted between the two organisms [68]. However, coccidioidomycosis
is extremely rare in California, suggesting that cats may be less susceptible to C.
immitis infections, or that feline exposure occurs to a greater degree in areas where
C. posadasii resides [67].
Coccidioides spp. is present in soil as a mycelium which germinate to form
arthroconidia that are released and dispersed when the soil is disturbed. Infection
occurs following inhalation of these arthrospores but also rarely by direct inoculation of organisms into skin. Dissemination occurs when the immune system is
incapable of containing replication of the organism to the lungs.
Deep Fungal Diseases
313
VetBooks.ir
Clinical Features in Cats
The mean age of affected cats at diagnosis was 6.2 years in one study [1] and 9 years
in another, [68] with age ranging from 3 to 17 years. Female cats were overrepresented in one study with 12 of 17 cases being female [68]. There is no breed
predilection reported and immunosuppression does not seem to play a role in the
development of the disease [68]. Although having access to outdoors is likely a risk
factor, cats housed exclusively indoors can also develop coccidioidomycosis [68,
69]. The reported duration of clinical signs before diagnosis of coccidioidomycosis
in cats ranged from less than 1 week to 1 year, with up to 86% of the animals having
clinical signs for less than a month before diagnosis [1, 65, 68].
Cats are mostly diagnosed when the disease has disseminated, and dermatologic
signs are the most common presenting complaints in cats with disseminated disease.
Dermatological signs were seen in 56% of 48 cats [65]. Cutaneous lesions include
plaque-like nodules, nodules with draining tracts, alopecia, scarring and induration
with draining tracts, papules, pustules and lingual ulcerations [65, 68, 70]. Regional
lymphadenopathy may also be present. More than half of the population of cats with
dermatologic signs also had clinical signs of systemic illness such as fever, lethargy,
weight loss, anorexia, lameness, or cough [68].
Respiratory signs such as cough or tachypnoea were noted in only 25% of cats
with coccidioidomycosis [65]. However, lung involvement is probably more common since thoracic radiographs were not performed on many cats of this study and
lung infection was found in nearly all cases at necropsy in another study [66]. Hilar
lymphadenopathy, interstitial or mixed interstitial and bronchointerstitial pulmonary
patterns and rarely pleural thickening or effusion are found on thoracic radiographs
[65]. Other reported clinical signs include ocular signs such as chorioretinitis, anterior uveitis, retinal detachment, panophthalmitis; CNS signs (with intracranial or
spinal cord lesions) such as seizures, hyperesthesia, behavioural changes, pelvic
limb weakness and ataxia; and musculoskeletal signs such as lameness [65, 66, 69,
71, 72].
Restrictive pericarditis, pericardial effusion and right sided heart failure are
reported in dogs with coccidioidomycosis but have not been described in cats [66].
However, pericardial involvement was found at necropsy in 26% of cats with coccidioidomycosis despite a lack of clinical signs referable to heart disease in all cats
of the study [66].
Diagnostic Tests
In cats with coccidioidomycosis, laboratory abnormalities include nonregenerative
anaemia, leucocytosis, leukopenia, hypoalbuminemia and hyperglobulinemia [65,
72]. In contrast to humans, eosinophilia has not been reported in cats with coccidioidomycosis [73]. The sensitivity and specificity of serology for the diagnosis of coccidioidomycosis in cats is unknown. Most commercial laboratories perform AGID
assays for immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies. In 39
VetBooks.ir
314
J. D. Lemetayer and J. E. Sykes
cats with coccidioidomycosis, all cats were seropositive at some point during their
illness [65]. At the time of diagnosis, 29 cats were positive for IgM (tube precipitin)
antibodies and six cats were negative for IgM but positive for IgG (complement
fixating) antibodies. IgG antibody titres ranged from 1:2 to 1:128, with 31 cats having titres ≥1:16.
Cytologic confirmation may be made by evaluation of aspirates of affected lymph
nodes, skin lesions, lungs, pleural effusion, as well as bronchoalveolar lavage but
is relatively insensitive for the diagnosis of coccidioidomycosis when compared
with other deep mycoses [67]. On cytology, a granulomatous or pyogranulomatous
inflammation is often seen, with sometimes rare multinucleated giant cells, few
eosinophils, and/or reactive lymphocytes. Occasionally a suppurative inflammatory
response predominates. If seen, the organism appears as a large (10–80 μm) round,
deeply basophilic, double-walled spherule that may contain endospores. The endospores are 2–5 μm in diameter, are surrounded by a thin, non-staining halo, and have
small, round to oval, densely aggregated, eccentric nuclei. Diff-Quik and Wright
stains can be used to facilitate visualization.
Similarly, multiple biopsy samples may need to be evaluated to identify the
organism histologically. The use of special stains such as PAS or GMS stains may
be required.
Coccidioides spp. structures were seen on cytology or histology of exudates or
tissue specimens in only 56% of 48 cats [65]. Cultures of exudates or tissues grew
Coccidioides in only 23% of these cats and therefore a negative culture does not
rule out a diagnosis of coccidioidomycosis. Coccidioides spp. can be isolated on
routine fungal media, but growth in culture represents a serious health hazard to
laboratory personnel and should only be performed if necessary and by suitably
equipped laboratories.
The use of PCR assays to aid in the diagnosis of coccidioidomycosis has not
been reported in cats.
Treatment and Prognosis
Itraconazole or fluconazole are typically used for the treatment of feline coccidioidomycosis, but ketoconazole has been used historically. Of 53 cats diagnosed with
coccidioidomycosis and treated mostly with ketoconazole, 67% survived with treatment [1]. The average treatment duration was 10 months.
Fluconazole may be used for patients with ocular and CNS involvement, because
of its superior penetration of the eyes and CNS [71, 72]. Itraconazole is preferred for
animals with bone involvement and is recommended for animals that fail to respond
to treatment with fluconazole. In people, a trend toward slightly greater efficacy
of itraconazole over fluconazole was found in non-meningeal cases but it was not
statistically significant [74]. In addition, the use of amphotericin B is recommended
for the treatment of human patients with very severe and/or rapidly progressing
VetBooks.ir
Deep Fungal Diseases
315
acute pulmonary or disseminated coccidioidomycosis, followed by fluconazole
once patients have stabilized [75].
In human patients who fail standard therapy, treatment with posaconazole or
voriconazole has been reported with approximately 70% of improvement and
slightly better outcomes with posaconazole compared to voriconazole [72, 76]. The
use of echinocandins in combination with voriconazole in refractory patients was
also reported with success [77]. However, voriconazole should be avoided in cats
due to their susceptibility to severe voriconazole toxicity.
Box 2: Mould Infections
• Mould infections are less prevalent in cats than in dogs.
• Cutaneous signs are rare with aspergillosis in cats. Aspergillus spp. cause
sino-nasal aspergillosis and sino-orbital aspergillosis in cats with a predisposition for brachycephalic cats. They also rarely cause disseminated disease, which is usually seen in immunodeficient cats.
• Hyalohyphomycosis and phaeohyphomycosis are cutaneous and subcutaneous infections usually acquired from traumatic implantation of fungi
from the environment. Most cats are immunocompetent.
• Zygomycosis is acquired by inhalation, ingestion or contamination of
wounds. Concurrent immunosuppressive conditions are common.
• Pythiosis is caused by the penetration of acquired motile biflagellate zoospores in aquatic environments through damaged skin or GI mucosa. It is
mostly manifested as cutaneous and subcutaneous lesions in cats.
• Complete surgical excision of the affected tissue with wide margins is the
treatment of choice for hyalohyphomycosis, phaeohyphomycosis, zygomycosis and pythiosis as treatment with anti-fungal drugs is usually not
curative.
Aspergillosis
Epidemiology
Aspergillus species are ubiquitous saprophytic moulds that are found worldwide in
soil and decaying vegetation [78]. Species affecting cats are usually included in the
A. fumigatus complex (Aspergillus fumigatus, Neosartorya spp., Aspergillus lentulus and Aspergillus udagawae) [79]. Members of the A. fumigatus complex cannot
be identified reliably by phenotypic testing alone and require molecular techniques
for identification [79]. Aspergillus flavus, Aspergillus nidulans, Aspergillus niger,
Aspergillus terreus, Aspergillus udagawae and Aspergillus felis have also been
detected in a few cases [78, 80, 81].
316
J. D. Lemetayer and J. E. Sykes
VetBooks.ir
Clinical Features in Cats
The most common forms of aspergillosis in cats are sino-nasal aspergillosis (SNA)
and sino-orbital aspergillosis (SOA). The development of localized sino-nasal or
sino-orbital infection suggest defects of local defence mechanisms. In normal conditions, infections are prevented by physical barriers such as mucociliary clearance, and the local innate immune system (macrophages and neutrophils) [82].
Brachycephalic breeds, especially Persian or Himalayan cats, are predisposed [78].
It has been suggested that this may result from reduced mucociliary clearance [78].
Other possible risk factors include previous viral upper respiratory tract infections,
inflammatory rhinitis and use of glucocorticoids or less likely previous antibiotic
treatment [80, 82]. No association between aspergillosis and feline retrovirus infections has been reported [79]. Affected cats range in age from 1.5 to 13 years (median,
5 years) with no clear sex predisposition [79]. The duration of clinical signs before
diagnosis ranged from less than 5 days to more than 6 weeks in one study [ 1].
Clinical signs of SNA include sneezing, uni- or bilateral serous to mucopurulent
nasal discharge, and sometimes epistaxis and less commonly stertorous breathing,
granuloma formation, soft tissue masses protruding from the nares, and bone lysis.
SOA is a more invasive form of SNA, with involvement of the retrobulbar space.
Clinical signs include unilateral exophthalmos, third eyelid prolapse, conjunctival hyperaemia and keratitis. With severe retrobulbar involvement, a mass may be
observed in the caudal aspect of the oral cavity (Fig. 5). CNS involvement, regional
lymphadenopathy and fever have also been described.
Systemic aspergillosis in cats is rare and is usually associated with immune-­
deficiency. Aspergillus niger pneumonia was reported in two cats with diabetes mellitus [83], and disseminated aspergillosis in 38 cats with more than half of these cats
Fig. 5 Mass in the caudal
aspect of the oral cavity of
a cat with sino-orbital
aspergillosis
VetBooks.ir
Deep Fungal Diseases
317
having a concurrent immune-suppressive condition (mainly panleukopenia, FeLV
and feline infectious peritonitis) [1, 84].
Cutaneous disease is also a very uncommon manifestation of aspergillosis in
cats. Cutaneous involvement of the naso-ocular region of a cat with naso-sinusal
aspergillosis was reported [85] and Aspergillus vitricola was cultured from an auricular lesion in another cat [86].
Diagnostic Tests
The diagnosis of SNA and SOA requires a combination of tests such as imaging
studies, rhinoscopy, cytology and/or histology, and fungal cultures. Imaging modalities such as computerized tomography (CT) scan or magnetic resonance imaging
(MRI) of the head can be used to evaluate for destruction of the nasal turbinates,
nasal septum, cribriform plate, and involvement of the sinus and retrobulbar space.
On rhinoscopy, destruction of the nasal turbinates and white-grey plaques may be
seen [87].
Cytologic examination of blind or rhinoscopy-directed mucosal swabs, brush
specimens of the nasal cavity, nasal biopsies from cats with SNA, or cytologic
examination of ultrasound- or CT-guided aspirates of retrobulbar masses from cats
with SOA often reveal mixed, predominantly pyogranulomatous inflammation.
Sometimes, Aspergillus hyphae are seen but false-negative results are common.
Aspergillus fumigatus can usually grow in few days to few weeks on routine
laboratory media and does not represent a significant hazard to laboratory personnel. In the absence of supportive rhinoscopic, cytologic or histopathologic findings, positive cultures from the nasal cavity require cautious interpretation because
Aspergillus spp. are ubiquitous and therefore false positive results are not uncommon. Whenever possible, fungal culture should be submitted from samples collected
by rhinoscopic guidance in order to increase sensitivity [88]. Growth of Aspergillus
from aspirates or biopsy specimens from a normally sterile site such as a retrobulbar mass strongly suggests a diagnosis of SOA. In one study, fungal cultures were
positive in 22/23 cats with SNA or SOA [79] but in another study the sensitivity of
culture was lower. [89] The use of serologic tests (antibody and antigen tests) for
diagnosis of aspergillosis in cats has been unreliable [78, 79, 81, 87, 89].
Treatment and Prognosis
Treatment of SNA in cats has been similar to treatment used in dogs. Intranasal
infusion of clotrimazole for 1 hour was described in three cats with good outcomes
[82, 87]. Treatment of SOA and disseminated aspergillosis requires systemic anti-­
fungal treatment (monotherapy or a combination of two anti-fungal treatments) but
prognosis is guarded to poor. Anti-fungal drugs reported in the treatment of SOA
and disseminated aspergillosis include itraconazole, amphotericin B, posaconazole,
voriconazole, terbinafine, caspofungin and micafungin [79, 90–92]. Voriconazole is
VetBooks.ir
318
J. D. Lemetayer and J. E. Sykes
not recommended because of the potential for severe toxicity in cats. Fluconazole
and flucytosine are not recommended because Aspergillus species are intrinsically
resistant to these anti-fungal drugs [93]. In addition, high minimum inhibitory concentrations (MIC) for ketoconazole are common among Aspergillus species.
In a 2015 Australian study evaluating anti-fungal resistance in canine and feline
isolates of Aspergillus fumigatus, the vast majority of isolates had low MICs for
itraconazole, voriconazole, posaconazole, clotrimazole and enilconazole [93].
Interestingly, seven isolates had high MICs for amphotericin B.
Aspergillus felis exhibits high MICs to many anti-fungal drugs [94]. High MICs
of A. felis isolates to at least one of the triazoles, and cross resistance among several
triazoles were observed. In addition, a high MIC for caspofungin was described for
one isolate.
Other Moulds
Hyalohyphomycosis
Hyalohyphomycosis is caused by non-dematiaceous (hyaline, nonpigmented) moulds.
A retrospective study from the UK evaluating 77 cats with nodular granulomatous
skin lesions caused by fungi found that the most frequent pathogens were hyalohyphomycetes [95]. Reported species associated with disease in cats include Fusarium,
Acremonium, Paecilomyces spp. and Metarhizium spp., among others [95–101].
These are filamentous fungi found in soil and on plants and have a worldwide
distribution.
Hyalohyphomycosis has been diagnosed in cats with cutaneous nodules, rhinosinusitis, pneumonia, pododermatitis and keratitis.
Diagnosis is made by cytology, histology and fungal culture. Cytological and
histological examination usually reveals pyogranulomatous inflammation in association with nonpigmented, frequently septate, branching hyphae that are often
pleomorphic. Culture and proper identification of the pathogen is recommended to
guide in the choice of anti-fungal treatment because some species are predictably
less susceptible to conventional anti-fungal drugs. However, because these fungi are
common laboratory contaminants and can sometimes be isolated from the skin or
hair of healthy animals, positive cultures from non-sterile sites should be considered
in light of the clinical picture.
Complete surgical excision of the affected tissue with wide margins is the treatment of choice whenever possible, followed by anti-fungal therapy for 3–6 months.
Drugs used most often to treat hyalohyphomycosis in small animals include
itraconazole and amphotericin B, but different fungal species vary in their
susceptibility to anti-fungal drugs. Posaconazole and the echinocandins, such
as caspofungin, may be more active against these fungi than itraconazole.
Fusarium spp. are intrinsically resistant to glucan synthesis inhibitors such as
caspofungin; however, in combination with amphotericin B, they can have a
synergistic action [98].
Deep Fungal Diseases
319
VetBooks.ir
Phaeohyphomycosis
Phaeohyphomycetes are dematiaceous filamentous fungi that contain a melanin-­
like pigment in the walls of the hyphae and occasionally cause opportunistic
infections in cats. The pigment plays important role in virulence and pathogenicity of these pathogens as it aids fungal evasion of the host immune response by
preventing hydrolytic enzymatic attack and scavenging of free radicals liberated
by phagocytic cells during then oxidative burst [102]. Species that have caused
disease in cats include Exophiala spp., Alternaria spp., Cladosporium spp.,
Phialophora spp., Cladophialophora spp., Ulocladium spp., Microsphaeropsis
spp., Fonsecae spp., Moniliella spp. and Aureobasidium spp., among others [86, 98, 102–115]. Cladosporium spp. may be more likely to disseminate
in immunocompetent cats. In addition, among the genus Cladophialophora,
Cladophialophora bantiana shows a marked neurotropism in comparison with
other fungi. [109, 115, 116]
Phaeohyphomycetes are found in soil, wood and decomposing plant debris
worldwide. Infections usually result from cutaneous inoculation resulting in cutaneous and subcutaneous infections (Fig. 6). Most lesions in cats occur on the head
or extremities and usually a single nodule is present. Systemic signs of illness are
usually absent. Rarely, ingestion or inhalation of spores might also occur and cause
deep infection [109].
Factors predisposing cats to phaeohyphomycoses may include treatment with
immunosuppressive agents; concurrent disease; or age-related, non-specific loss of
immunity. However, no obvious immunosuppression is found in most cases [103]
although the majority of cats in reports were not tested for FIV and FeLV.
Diagnosis is made by cytology, histology and fungal culture. Cytology of exudates usually reveals pyogranulomatous inflammation that may contain pigmented
fungal hyphae, pseudohyphae, and/or yeast-like cells. Fungal culture is recommended for proper diagnosis. An indirect ELISA has been developed for the
Fig. 6 Subcutaneous
phaeohyphomycosis
associated with swelling of
the distal limb of a cat
VetBooks.ir
320
J. D. Lemetayer and J. E. Sykes
detection of anti-Alternaria IgG antibodies in serum in domestic cats. However, cats
with disease caused by Alternaria did not have significantly higher concentrations
of antibody than healthy cats or cats with other diseases [117].
Complete surgical excision of the affected tissue with wide margins is the treatment of choice whenever possible, followed by anti-fungal therapy for 3 to 6 months.
If complete surgical removal is not possible, the prognosis is guarded. Indeed, phaeohyphomycoses often have recrudescent clinical courses and are refractory to
many anti-fungal drugs. In cases of disseminated or cerebral infection, treatment is
rarely successful, and prognosis is poor. Ketoconazole, itraconazole, amphotericin
B, flucytosine and terbinafine have been used with variable results to treat phaeohyphomycosis in cats [111, 113]. Combination therapy with terbinafine and an azole
anti-fungal drug such as itraconazole or posaconazole has been suggested [114].
If improvement is seen, long-term treatment (6 to 12 months) is recommended to
prevent recurrence of the lesions.
Zygomycosis
Zygomycetes are opportunistic organisms present in the soil, water, decaying matter and faeces. They include organisms that belong to the genera Basidiobolus and
Conidiobolus in the order Entomophthorales, and the genera Rhizopus, Absidia,
Mucor, Saksenaea, and others in the order Mucorales [118]. Infection is believed to
be acquired by inhalation, ingestion or contamination of wounds. Rare reports exist
of Mucor spp. infections in cats, including a case of cerebral mycosis, subcutaneous
infection and duodenal perforation caused by Rhizomucor spp. [119–121] In addition, 12 cases of suspected mucormycosis diagnosed using histology were reported
in a necropsy study [84]. Lesions in most of these cats involved the GI tract or lungs
and 6 of the 12 cats had possible immune-suppressive conditions.
Conidiobolus infection was suspected in a 3-year-old cat with an ulcerative
lesion of the hard palate [118].
A definitive diagnosis of zygomycosis is made based on cytological or histopathological examination in combination with fungal culture. Cytological and histological findings include pyogranulomatous, suppurative or eosinophilic inflammation.
Broad (>8 μm), poorly septate hyphae with thick prominent eosinophilic sleeves are
sometimes observed. Microscopic examination of macerated tissue that has been
digested in 10% potassium hydroxide may be more likely to reveal the hyphal elements. Staining of histopathological specimens with GMS and PAS stains can also
assist in visualization of hyphal elements.
Wide surgical excision (whenever possible) combined with long-term medical
treatment is recommended for zygomycosis. Zygomycetes are variably susceptible to anti-fungal drugs. Posaconazole and amphotericin B are considered the
most effective anti-fungal drugs for Mucor infections in humans [120]. Some
cases of human zygomycosis also had good outcome with itraconazole treatment
[122, 123].
Deep Fungal Diseases
321
VetBooks.ir
Pythiosis
Pythiosis is caused by the aquatic oomycete Pythium insidiosum. Oomycetes are
soil and aquatic organisms that are phylogenetically distant from the fungi, and
more closely related to algae [118]. Chitin, an essential component of the fungal
cell wall, is generally lacking in the oomycete cell wall, which instead contains
predominately cellulose and β–glucan [118]. Ergosterol is also not a principal sterol
in the oomycete cell membrane contrary to fungal organisms.
The infective form of P. insidiosum is a motile biflagellate zoospore, which is
released into aquatic environments and causes infection by penetrating damaged
skin or GI mucosa. Pythiosis is most commonly encountered in tropical and subtropical climates; however, infections in animals from temperate areas have also
been reported [124]. It is endemic in the USA (primarily in the Gulf coast states),
and cases also occur in Southeast Asia, eastern coastal Australia, New Zealand, and
South America [118].
Pythiosis is extremely rare in cats and usually manifests as subcutaneous lesions
(including in the inguinal, tail head, or periorbital regions), draining nodular lesions
or ulcerated plaque-like lesions localized on the extremities [118]. One cat with
nasal and retrobulbar mass [125], one cat with a sublingual mass [126] and two cats
with gastro-intestinal pythiosis [124] were also reported. Specific breed and sex
predilections have not been observed but young cats may be predisposed. In 10 cats
with cutaneous lesions caused by P. insidiosum, five were younger than 10 months
old, with an age range of 4 months to 9 years [118].
Cytological and histological examinations show eosinophilic and granulomatous
inflammation with prominent fibrosis and necrosis [124]. To complicate the diagnosis, P. insidiosum typically does not stain with H&E stain and may be present in low
numbers. The hyphae appear within necrotic areas and granulomas as clear round
or oval to elongate structures delineated by a narrow rim of eosinophilic material.
It also stains poorly with PAS but can be observed with GMS stain. The hyphae
are infrequently septate, branching and measure from 2.5 to 8.9 mm in diameter
with thick walls in comparison to the septa and with almost parallel sides [124].
Differentiation between pythiosis, lagenidiosis, and zygomycosis based on routine
histologic examination is not usually possible because differences in their histologic
characteristics are subtle, although Mucor spp. stain equally well using H&E, PAS,
and GMS stains.
Culture of tissues or the use of immunohistochemistry, PCR and/or serology
can aid in diagnosis [124]. Culture of exudates is usually unsuccessful and culture
of tissues requires specific specimen handling (unrefrigerated tissues that are kept
moist) and culture techniques. The identity of organisms isolated in culture can be
confirmed using PCR sequencing [127].
In addition, immunoblot serology and ELISA techniques have been used successfully to support the diagnosis of pythiosis in a few cats [124, 125, 128].
The treatment of choice for pythiosis is aggressive surgical resection of infected
tissues with 3–4-cm margins whenever possible. When used alone, medical therapy
VetBooks.ir
322
J. D. Lemetayer and J. E. Sykes
for pythiosis is typically unrewarding. This is likely because ergosterol, which is the
target for most anti-fungal drugs, is generally lacking in the oomycete cell membrane. In dogs, a combination of itraconazole and terbinafine may be effective for
resolution of incompletely resected or nonresectable lesions. Ketoconazole has also
been used [125]. The short-term use of prednisone is also recommended in dogs
with gastrointestinal pythiosis to improve clinical signs (vomiting, decreased appetite) [127].
References
1. Davies C, Troy GC. Deep mycotic infections in cats. J Am Anim Hosp Assoc. 1996;32:380–91.
2. Sykes JE, Taboada J. Histoplasmosis. In: Sykes JE, editor. Canine and feline infectious diseases. St Louis: Elsevier Saunders; 2014. p. 587–98.
3. Kano R, Hosaka S, Hasegawa A. First isolation of Cryptococcus magnus from a cat.
Mycopathologia. 2004;157:263–4.
4. Poth T, Seibold M, Werckenthin C, Hermanns W. First report of a Cryptococcus magnus infection in a cat. Med Mycol. 2010;48:1000–4.
5. Kano R, Kitagawat M, Oota S, Oosumi T, Murakami Y, Tokuriki M, et al. First case of feline
systemic Cryptococcus albidus infection. Med Mycol. 2008;46:75–7.
6. O’Brien CR, Krockenberger MB, Wigney DI, Martin P, Malik R. Retrospective study of
feline and canine cryptococcosis in Australia from 1981 to 2001: 195 cases. Med Mycol.
2004;42:449–60.
7. Lester SJ, Malik R, Bartlett KH, Duncan CG. Cryptococcosis: update and emergence of
Cryptococcus gattii. Vet Clin Pathol. 2011;40:4–17.
8. Pennisi MG, Hartmann K, Lloret A, Ferrer L, Addie D, Belak S, et al. Cryptococcosis in cats:
ABCD guidelines on prevention and management. J Feline Med Surg. 2013;15:611–8.
9. McGill S, Malik R, Saul N, Beetson S, Secombe C, Robertson I, et al. Cryptococcosis in
domestic animals in Western Australia: a retrospective study from 1995–2006. Med Mycol.
2009;47:625–39.
10. Malik R, Wigney DI, Muir DB, Gregory DJ, Love DN. Cryptococcosis in cats: clinical and
mycological assessment of 29 cases and evaluation of treatment using orally administered
fluconazole. J Med Vet Mycol. 1992;30:133–44.
11. Trivedi SR, Sykes JE, Cannon MS, Wisner ER, Meyer W, Sturges BK, et al. Clinical features
and epidemiology of cryptococcosis in cats and dogs in California: 93 cases (1988–2010). J
Am Vet Med Assoc. 2011;239:357–69.
12. Jacobs GJ, Medleau L, Calvert C, Brown J. Cryptococcal infection in cats: factors influencing
treatment outcome, and results of sequential serum antigen titers in 35 cats. J Vet Intern Med.
1997;11:1–4.
13. Castrodale LJ, Gerlach RF, Preziosi DE, Frederickson P, Lockhart SR. Prolonged incubation period for Cryptococcus gattii infection in cat, Alaska, USA. Emerg Infect Dis.
2013;19:1034–5.
14. Myers A, Meason-Smith C, Mansell J, Krockenberger M, Peters-Kennedy J, Ross Payne H, et al.
Atypical cutaneous cryptococcosis in four cats in the USA. Vet Dermatol. 2017;28:405–e97.
15. Odom T, Anderson JG. Proliferative gingival lesion in a cat with disseminated cryptococcosis.
J Vet Dent. 2000;17:177–81.
16. Siak MK, Paul A, Drees R, Arthur I, Burrows AK, Tebb AJ, et al. Otogenic meningoencephalomyelitis due to Cryptococcus gattii (VGII) infection in a cat from Western Australia. JFMS
Open Rep. 2015;1:2055116915585022.
17. Sykes JE, Malik R. Cryptococcosis. In: Sykes JE, editor. Canine and feline infectious diseases.
St Louis: Elsevier Saunders; 2014. p. 599–612.
VetBooks.ir
Deep Fungal Diseases
323
18. Trivedi SR, Malik R, Meyer W, Sykes JE. Feline cryptococcosis: impact of current research on
clinical management. J Feline Med Surg. 2011;13:163–72.
19. Guess T, Lai H, Smith SE, Sircy L, Cunningham K, Nelson DE, et al. Size matters: measurement of capsule diameter in Cryptococcus neoformans. J Vis Exp. 2018;132:1–10.
20. Ranjan R, Jain D, Singh L, Iyer VK, Sharma MC, Mathur SR. Differentiation of histoplasma
and cryptococcus in cytology smears: a diagnostic dilemma in severely necrotic cases.
Cytopathology. 2015;26:244–9.
21. Sykes JE, Hodge G, Singapuri A, Yang ML, Gelli A, Thompson GR 3rd. In vivo development of fluconazole resistance in serial Cryptococcus gattii isolates from a cat. Med Mycol.
2017;55:396–401.
22. Kano R, Okubo M, Yanai T, Hasegawa A, Kamata H. First isolation of azole-resistant
Cryptococcus neoformans from feline cryptococcosis. Mycopathologia. 2015;180:427–33.
23. Mondon P, Petter R, Amalfitano G, Luzzati R, Concia E, Polacheck I, et al. Heteroresistance
to fluconazole and voriconazole in Cryptococcus neoformans. Antimicrob Agents Chemother.
1999;43:1856–61.
24. O’Brien CR, Krockenberger MB, Martin P, Wigney DI, Malik R. Long-term outcome of therapy for 59 cats and 11 dogs with cryptococcosis. Aust Vet J. 2006;84:384–92.
25. Sykes JE, Sturges BK, Cannon MS, Gericota B, Higgins RJ, Trivedi SR, et al. Clinical signs,
imaging features, neuropathology, and outcome in cats and dogs with central nervous system
cryptococcosis from California. J Vet Intern Med. 2010;24:1427–38.
26. Barrs VR, Martin P, Nicoll RG, Beatty JA, Malik R. Pulmonary cryptococcosis and Capillaria
aerophila infection in an FIV-positive cat. Aust Vet J. 2000;78:154–8.
27. Balajee SA, Hurst SF, Chang LS, Miles M, Beeler E, Hale C, et al. Multilocus sequence typing
of Histoplasma capsulatum in formalin-fixed paraffin-embedded tissues from cats living in
non-endemic regions reveals a new phylogenetic clade. Med Mycol. 2013;51:345–51.
28. Fischer NM, Favrot C, Monod M, Grest P, Rech K, Wilhelm S. A case in Europe of feline
histoplasmosis apparently limited to the skin. Vet Dermatol. 2013;24:635–8.
29. Percy DH. Feline histoplasmosis with ocular involvement. Vet Pathol. 1981;18:163–9.
30. Larsuprom L, Duangkaew L, Kasorndorkbua C, Chen C, Chindamporn A, Worasilchai
N. Feline cutaneous histoplasmosis: the first case report from Thailand. Med Mycol Case Rep.
2017;18:28–30.
31. Mavropoulou A, Grandi G, Calvi L, Passeri B, Volta A, Kramer LH, et al. Disseminated histoplasmosis in a cat in Europe. J Small Anim Pract. 2010;51:176–80.
32. Kasuga T, White TJ, Koenig G, McEwen J, Restrepo A, Castaneda E, et al. Phylogeography of
the fungal pathogen Histoplasma capsulatum. Mol Ecol. 2003;12:3383–401.
33. Bromel C, Sykes JE. Histoplasmosis in dogs and cats. Clin Tech Small Anim Pract.
2005;20:227–32.
34. Aulakh HK, Aulakh KS, Troy GC. Feline histoplasmosis: a retrospective study of 22 cases
(1986–2009). J Am Anim Hosp Assoc. 2012;48:182–7.
35. Reinhart JM, KuKanich KS, Jackson T, Harkin KR. Feline histoplasmosis: fluconazole therapy
and identification of potential sources of Histoplasma species exposure. J Feline Med Surg.
2012;14:841–8.
36. Atiee G, Kvitko-White H, Spaulding K, Johnson M. Ultrasonographic appearance of histoplasmosis identified in the spleen in 15 cats. Vet Radiol Ultrasoun. 2014;55:310–4.
37. Gingerich K, Guptill L. Canine and feline histoplasmosis: a review of a widespread fungus. Vet
Med. 2008;103:248–64.
38. Clinkenbeard KD, Cowell RL, Tyler RD. Disseminated histoplasmosis in cats: 12 cases
(1981–1986). J Am Vet Med Assoc. 1987;190:1445–8.
39. Wolf AM. Histoplasma capsulatum osteomyelitis in the cat. J Vet Intern Med. 1987;1:158–62.
40. Carneiro RA, Lavalle GE, Araujo RB. Cutaneous histoplasmosis in cat: a case report. Arq Bras
Med Vet Zoo. 2005;57:158–61.
41. Tamulevicus AM, Harkin K, Janardhan K, Debey BM. Disseminated histoplasmosis accompanied by cutaneous fragility in a cat. J Am Anim Hosp Assoc. 2011;47:E36–41.
VetBooks.ir
324
J. D. Lemetayer and J. E. Sykes
42. Vinayak A, Kerwin SC, Pool RR. Treatment of thoracolumbar spinal cord compression associated with Histoplasma capsulatum infection in a cat. J Am Vet Med Assoc. 2007;230:1018–23.
43. Lamm CG, Rizzi TE, Campbell GA, Brunker JD. Pathology in practice. Histoplasma capsulatum Infections. J Am Vet Med Assoc. 2009;235:155–7.
44. Taylor AR, Barr JW, Hokamp JA, Johnson MC, Young BD. Cytologic diagnosis of disseminated histoplasmosis in the wall of the urinary bladder of a cat. J Am Anim Hosp Assoc.
2012;48:203–8.
45. Hodges RD, Legendre AM, Adams LG, Willard MD, Pitts RP, Monce K, et al. Itraconazole for
the treatment of histoplasmosis in cats. J Vet Intern Med. 1994;8:409–13.
46. Hanzlicek AS, Meinkoth JH, Renschler JS, Goad C, Wheat LJ. Antigen concentrations as an
indicator of clinical remission and disease relapse in cats with histoplasmosis. J Vet Intern
Med. 2016;30:1065–73.
47. Cook AK, Cunningham LY, Cowell AK, Wheat LJ. Clinical evaluation of urine Histoplasma
capsulatum antigen measurement in cats with suspected disseminated histoplasmosis. J Feline
Med Surg. 2012;14:512–5.
48. Klang A, Loncaric I, Spergser J, Eigelsreiter S, Weissenbock H. Disseminated histoplasmosis
in a domestic cat imported from the USA to Austria. Med Mycol Case Rep. 2013;2:108–12.
49. Spec A, Connoly P, Montejano R, Wheat LJ. In vitro activity of isavuconazole against
fluconazole-­resistant isolates of Histoplasma capsulatum. Med Mycol. 2018;56:834–7.
50. Renschler JS, Norsworthy GD, Rakian RA, Rakian AI, Wheat LJ, Hanzlicek
AS. Reduced susceptibility to fluconazole in a cat with histoplasmosis. JFMS Open Rep.
2017;3:2055116917743364.
51. Bromel C, Sykes JE. Epidemiology, diagnosis, and treatment of blastomycosis in dogs and
cats. Clin Tech Small Anim Pract. 2005;20:233–9.
52. Gilor C, Graves TK, Barger AM, O’Dell-Anderson K. Clinical aspects of natural infection with
Blastomyces dermatitidis in cats: 8 cases (1991–2005). J Am Vet Med Assoc. 2006;229:96–9.
53. Davies JL, Epp T, Burgess HJ. Prevalence and geographic distribution of canine and feline
blastomycosis in the Canadian prairies. Can Vet J. 2013;54:753–60.
54. Easton KL. Cutaneous North American blastomycosis in a Siamese cat. Can Vet J.
1961;2:350–1.
55. Duangkaew L, Larsuprom L, Kasondorkbua C, Chen C, Chindamporn A. Cutaneous blastomycosis and dermatophytic pseudomycetoma in a Persian cat from Bangkok, Thailand. Med
Mycol Case Rep. 2017;15:12–5.
56. Miller PE, Miller LM, Schoster JV. Feline blastomycosis – a report of 3 cases and literature-­
review (1961 to 1988). J Am Anim Hosp Assoc. 1990;26:417–24.
57. Blondin N, Baumgardner DJ, Moore GE, Glickman LT. Blastomycosis in indoor cats: suburban Chicago, Illinois, USA. Mycopathologia. 2007;163:59–66.
58. Houseright RA, Webb JL, Claus KN. Pathology in practice. Blastomycosis in an indoor-only
cat. J Am Vet Med Assoc. 2015;247:357–9.
59. Stern JA, Chew DJ, Schissler JR, Green EM. Cutaneous and systemic blastomycosis, hypercalcemia, and excess synthesis of calcitriol in a domestic shorthair cat. J Am Anim Hosp
Assoc. 2011;47:e116–20.
60. Meece JK, Anderson JL, Klein BS, Sullivan TD, Foley SL, Baumgardner DJ, et al. Genetic
diversity in Blastomyces dermatitidis: implications for PCR detection in clinical and environmental samples. Med Mycol. 2010;48:285–90.
61. Sidamonidze K, Peck MK, Perez M, Baumgardner D, Smith G, Chaturvedi V, et al. Real-­
time PCR assay for identification of Blastomyces dermatitidis in culture and in tissue. J Clin
Microbiol. 2012;50:1783–6.
62. Smith JR, Legendre AM, Thomas WB, LeBlanc CJ, Lamkin C, Avenell JS, et al. Cerebral
Blastomyces dermatitidis infection in a cat. J Am Vet Med Assoc. 2007;231:1210–4.
63. Breider MA, Walker TL, Legendre AM, VanEe RT. Blastomycosis in cats: five cases (1979–
1986). J Am Vet Med Assoc. 1988;193:570–2.
64. McBride JA, Gauthier GM, Klein BS. Clinical manifestations and treatment of blastomycosis.
Clin Chest Med. 2017;38:435–49.
VetBooks.ir
Deep Fungal Diseases
325
65. Greene RT, Troy GC. Coccidioidomycosis in 48 cats – a retrospective study (1984–1993). J
Vet Intern Med. 1995;9:86–91.
66. Graupmann-Kuzma A, Valentine BA, Shubitz LF, Dial SM, Watrous B, Tornquist
SJ. Coccidioidomycosis in dogs and cats: a review. J Am Anim Hosp Assoc. 2008;44:226–35.
67. Sykes JE. Coccidioidomycosis. In: Sykes JE, editor. Canine and feline infectious diseases. St
Louis: Elsevier Saunders; 2014. p. 613–23.
68. Simoes DM, Dial SM, Coyner KS, Schick AE, Lewis TP. Retrospective analysis of cutaneous lesions in 23 canine and 17 feline cases of coccidiodomycosis seen in Arizona, USA
(2009–2015). Vet Dermatol. 2016;27:346.
69. Foureman P, Longshore R, Plummer SB. Spinal cord granuloma due to Coccidioides immitis
in a cat. J Vet Intern Med. 2005;19:373–6.
70. Amorim I, Colimao MJ, Cortez PP, Dias Pereira P. Coccidioidomycosis in a cat imported from
the USA to Portugal. Vet Rec. 2011;169: 232a.
71. Bentley RT, Heng HG, Thompson C, Lee CS, Kroll RA, Roy ME, et al. Magnetic resonance
imaging features and outcome for solitary central nervous system Coccidioides granulomas in
11 dogs and cats. Vet Radiol Ultrasoun. 2015;56:520–30.
72. Tofflemire K, Betbeze C. Three cases of feline ocular coccidioidomycosis: presentation, clinical features, diagnosis, and treatment. Vet Ophthalmol. 2010;13:166–72.
73. Alzoubaidi MSS, Knox KS, Wolk DM, Nesbit LA, Jahan K, Luraschi-Monjagatta
C. Eosinophilia in coccidioidomycosis. Am J Resp Crit Care. 2013;187
74. Galgiani JN, Catanzaro A, Cloud GA, Johnson RH, Williams PL, Mirels LF, et al. Comparison
of oral fluconazole and itraconazole for progressive, nonmeningeal coccidioidomycosis. A
randomized, double-blind trial. Mycoses Study Group. Ann Intern Med. 2000;133:676–86.
75. Galgiani JN, Ampel NM, Blair JE, Catanzaro A, Geertsma F, Hoover SE, et al. 2016 Infectious
Diseases Society of America (IDSA) Clinical Practice Guideline for the treatment of coccidioidomycosis. Clin Infect Dis. 2016;63:E112–E46.
76. Kim MM, Vikram HR, Kusne S, Seville MT, Blair JE. Treatment of refractory coccidioidomycosis with voriconazole or posaconazole. Clin Infect Dis. 2011;53:1060–6.
77. Levy ER, McCarty JM, Shane AL, Weintrub PS. Treatment of pediatric refractory coccidioidomycosis with combination voriconazole and caspofungin: a retrospective case series. Clin
Infect Dis. 2013;56:1573–8.
78. Hartmann K, Lloret A, Pennisi MG, Ferrer L, Addie D, Belak S, et al. Aspergillosis in cats:
ABCD guidelines on prevention and management. J Feline Med Surg. 2013;15:605–10.
79. Barrs VR, Halliday C, Martin P, Wilson B, Krockenberger M, Gunew M, et al. Sinonasal and
sino-orbital aspergillosis in 23 cats: aetiology, clinicopathological features and treatment outcomes. Vet J. 2012;191:58–64.
80. Barachetti L, Mortellaro CM, Di Giancamillo M, Giudice C, Martino P, Travetti O, et al.
Bilateral orbital and nasal aspergillosis in a cat. Vet Ophthalmol. 2009;12:176–82.
81. Whitney BL, Broussard J, Stefanacci JD. Four cats with fungal rhinitis. J Feline Med Surg.
2005;7:53–8.
82. Tomsa K, Glaus TA, Zimmer C, Greene CE. Fungal rhinitis and sinusitis in three cats. J Am
Vet Med Assoc. 2003;222:1380–4.
83. Leite RV, Fredo G, Lupion CG, Spanamberg A, Carvalho G, Ferreiro L, et al. Chronic invasive
pulmonary aspergillosis in two cats with diabetes mellitus. J Comp Pathol. 2016;155:141–4.
84. Ossent P. Systemic aspergillosis and mucormycosis in 23 cats. Vet Rec. 1987;120:330–3.
85. Malik R, Vogelnest L, O’Brien CR, White J, Hawke C, Wigney DI, et al. Infections and some
other conditions affecting the skin and subcutis of the naso-ocular region of cats--clinical
experience 1987–2003. J Feline Med Surg. 2004;6:383–90.
86. Bernhardt A, von Bomhard W, Antweiler E, Tintelnot K. Molecular identification of fungal
pathogens in nodular skin lesions of cats. Med Mycol. 2015;53:132–44.
87. Furrow E, Groman RP. Intranasal infusion of clotrimazole for the treatment of nasal aspergillosis in two cats. J Am Vet Med Assoc. 2009;235:1188–93.
88. Sykes JE. Aspergillosis. In: Sykes JE, editor. Canine and feline infectious diseases. St Louis:
Elsevier Saunders; 2014. p. 633–59.
VetBooks.ir
326
J. D. Lemetayer and J. E. Sykes
89. Goodall SA, Lane JG, Warnock DW. The diagnosis and treatment of a case of nasal aspergillosis in a cat. J Small Anim Pract. 1984;25:627–33.
90. McLellan GJ, Aquino SM, Mason DR, Kinyon JM, Myers RK. Use of posaconazole in the
management of invasive orbital aspergillosis in a cat. J Am Anim Hosp Assoc. 2006;42:302–7.
91. Smith LN, Hoffman SB. A case series of unilateral orbital aspergillosis in three cats and treatment with voriconazole. Vet Ophthalmol. 2010;13:190–203.
92. Kano R, Itamoto K, Okuda M, Inokuma H, Hasegawa A, Balajee SA. Isolation of Aspergillus
udagawae from a fatal case of feline orbital aspergillosis. Mycoses. 2008;51:360–1.
93. Talbot JJ, Kidd SE, Martin P, Beatty JA, Barrs VR. Azole resistance in canine and feline
isolates of Aspergillus fumigatus. Comp Immunol Microbiol Infect Dis. 2015;42:37–41.
94. Barrs VR, van Doorn TM, Houbraken J, Kidd SE, Martin P, Pinheiro MD, et al. Aspergillus
felis sp nov., an emerging agent of invasive aspergillosis in humans, cats, and dogs. PLoS
One. 2013;8:e64871.
95. Miller RI. Nodular granulomatous fungal skin diseases of cats in the United Kingdom: a
retrospective review. Vet Dermatol. 2010;21:130–5.
96. Leperlier D, Vallefuoco R, Laloy E, Debeaupuits J, Thibaud PD, Crespeau FL, et al.
Fungal rhinosinusitis caused by Scedosporium apiospermum in a cat. J Feline Med Surg.
2010;12:967–71.
97. Pawloski DR, Brunker JD, Singh K, Sutton DA. Pulmonary Paecilomyces lilacinus infection
in a cat. J Am Anim Hosp Assoc. 2010;46:197–202.
98. Kluger EK, Della Torre PK, Martin P, Krockenberger MB, Malik R. Concurrent Fusarium
chlamydosporum and Microsphaeropsis arundinis infections in a cat. J Feline Med Surg.
2004;6:271–7.
99. Sugahara G, Kiuchi A, Usui R, Usui R, Mineshige T, Kamiie J, et al. Granulomatous pododermatitis in the digits caused by Fusarium proliferatum in a cat. J Vet Med Sci. 2014;76:
435–8.
100. Binder DR, Sugrue JE, Herring IP. Acremonium keratomycosis in a cat. Vet Ophthalmol.
2011;14(Suppl 1):111–6.
101. Muir D, Martin P, Kendall K, Malik R. Invasive hyphomycotic rhinitis in a cat due to
Metarhizium anisopliae. Med Mycol. 1998;36:51–4.
102. Overy DP, Martin C, Muckle A, Lund L, Wood J, Hanna P. Cutaneous Phaeohyphomycosis
caused by Exophiala attenuata in a domestic cat. Mycopathologia. 2015;180:281–7.
103. Tennant K, Patterson-Kane J, Boag AK, Rycroft AN. Nasal mycosis in two cats caused by
Alternaria species. Vet Rec. 2004;155:368–70.
104. Bostock DE, Coloe PJ, Castellani A. Phaeohyphomycosis caused by Exophiala jeanselmei in
a domestic cat. J Comp Pathol. 1982;92:479.
105. Dion WM, Pukay BP, Bundza A. Feline Cutaneous phaeohyphomycosis caused by
Phialophora verrucosa. Can Vet J. 1982;23:48–9.
106. Sisk DB, Chandler FW. Phaeohyphomycosis and cryptococcosis in a cat. Vet Pathol.
1982;19:554–6.
107. Kettlewell P, McGinnis MR, Wilkinson GT. Phaeohyphomycosis caused by Exophiala spinifera in two cats. J Med Vet Mycol. 1989;27:257–64.
108. Nuttal W, Woodgyer A, Butler S. Phaeohyphomycosis caused by Exophiala jeanselmei in a
domestic cat. N Z Vet J. 1990;38:123.
109. Abramo F, Bastelli F, Nardoni S, Mancianti F. Feline cutaneous phaeohyphomycosis due to
Cladophyalophora bantiana. J Feline Med Surg. 2002;4:157–63.
110. Beccati M, Vercelli A, Peano A, Gallo MG. Phaeohyphomycosis by Phialophora verrucosa:
first European case in a cat. Vet Rec. 2005;157:93–4.
111. Knights CB, Lee K, Rycroft AN, Patterson-Kane JC, Baines SJ. Phaeohyphomycosis caused
by Ulocladium species in a cat. Vet Rec. 2008;162:415–6.
112. McKenzie RA, Connole MD, McGinnis MR, Lepelaar R. Subcutaneous phaeohyphomycosis
caused by Moniliella suaveolens in two cats. Vet Pathol. 1984;21:582–6.
113. Fondati A, Gallo MG, Romano E, Fondevila D. A case of feline phaeohyphomycosis due to
Fonsecaea pedrosoi. Vet Dermatol. 2001;12:297–301.
VetBooks.ir
Deep Fungal Diseases
327
114. Evans N, Gunew M, Marshall R, Martin P, Barrs V. Focal pulmonary granuloma caused by
Cladophialophora bantiana in a domestic short haired cat. Med Mycol. 2011;49:194–7.
115. Bouljihad M, Lindeman CJ, Hayden DW. Pyogranulomatous meningoencephalitis associated
with dematiaceous fungal (Cladophialophora bantiana) infection in a domestic cat. J Vet
Diagn Investig. 2002;14:70–2.
116. Lavely J, Lipsitz D. Fungal infections of the central nervous system in the dog and cat. Clin
Tech Small Anim Pract. 2005;20:212–9.
117. Dye C, Peters I, Tasker S, Caney SMA, Dye S, Gruffydd-Jones TJ, et al. Preliminary study
using an indirect ELISA for the detection of serum antibodies to Alternaria in domestic cats.
Vet Rec. 2005;156:633–5.
118. Grooters AM. Pythiosis, lagenidiosis, and zygomycosis in small animals. Vet Clin North Am
Small Anim Pract. 2003;33:695–720. v
119. Ravisse P, Fromentin H, Destombes P, Mariat F. Cerebral mucormycosis in the cat caused by
Mucor pusillus. Sabouraudia. 1978;16:291–8.
120. Wray JD, Sparkes AH, Johnson EM. Infection of the subcutis of the nose in a cat caused by
Mucor species: successful treatment using posaconazole. J Feline Med Surg. 2008;10:523–7.
121. Cunha SC, Aguero C, Damico CB, Corgozinho KB, Souza HJ, Pimenta AL, et al. Duodenal
perforation caused by Rhizomucor species in a cat. J Feline Med Surg. 2011;13:205–7.
122. Mahamaytakit N, Singalavanija S, Limpongsanurak W. Subcutaneous zygomycosis in children: 2 case reports. J Med Assoc Thail. 2014;97(Suppl 6):S248–53.
123. Eisen DP, Robson J. Complete resolution of pulmonary Rhizopus oryzae infection with
itraconazole treatment: more evidence of the utility of azoles for zygomycosis. Mycoses.
2004;47:159–62.
124. Rakich PM, Grooters AM, Tang KN. Gastrointestinal pythiosis in two cats. J Vet Diagn
Investig. 2005;17:262–9.
125. Bissonnette KW, Sharp NJ, Dykstra MH, Robertson IR, Davis B, Padhye AA, et al. Nasal and
retrobulbar mass in a cat caused by Pythium insidiosum. J Med Vet Mycol. 1991;29:39–44.
126. Fortin JS, Calcutt MJ, Kim DY. Sublingual pythiosis in a cat. Acta Vet Scand. 2017;59
127. Grooters AM. Pythiosis, lagenidiosis, and zygomycosis. In: Sykes JE, editor. Canine and
feline infectious diseases. St Louis: Elsevier Saunders; 2014. p. 668–78.
128. Thomas RC, Lewis DT. Pythiosis in dogs and cats. Comp Cont Educ Pract. 1998;20: 63.
129. Middleton SM, Kubier A, Dirikolu L, Papich MG, Mitchell MA, Rubin SI. Alternate-day
dosing of itraconazole in healthy adult cats. J Vet Pharmacol Ther. 2016;39:27–31.
130. Mawby DI, Whittemore JC, Fowler LE, Papich MG. Comparison of absorption characteristics of oral reference and compounded itraconazole formulations in healthy cats. J Am Vet
Med Assoc. 2018;252:195–200.
131. Vishkautsan P, Papich MG, Thompson GR 3rd, Sykes JE. Pharmacokinetics of voriconazole
after intravenous and oral administration to healthy cats. Am J Vet Res. 2016;77:931–9.
132. Mawby DI, Whittemore JC, Fowler LE, Papich MG. Posaconazole pharmacokinetics in
healthy cats after oral and intravenous administration. J Vet Intern Med. 2016;30:1703–7.
133. Vaden SL, Heit MC, Hawkins EC, Manaugh C, Riviere JE. Fluconazole in cats: pharmacokinetics following intravenous and oral administration and penetration into cerebrospinal fluid,
aqueous humour and pulmonary epithelial lining fluid. J Vet Pharmacol Ther. 1997;20:181–6.
134. Medleau L, Jacobs GJ, Marks MA. Itraconazole for the treatment of cryptococcosis in cats. J
Vet Intern Med. 1995;9:39–42.
135. Boothe DM, Herring I, Calvin J, Way N, Dvorak J. Itraconazole disposition after single oral
and intravenous and multiple oral dosing in healthy cats. Am J Vet Res. 1997;58:872–7.
136. Leshinsky J, McLachlan A, Foster DJR, Norris R, Barrs VR. Pharmacokinetics of caspofungin acetate to guide optimal dosing in cats. PLoS One. 2017;12:e0178783.
137. Ghannoum MA. Antifungal resistance: monitoring for terbinafine resistance among clinical
dermatophyte isolates. Mycoses. 2013;56:38.
138. Rocha EMF, Gardiner RE, Park S, Martinez-Rossi NM, Perlin DS. A Phe389Leu substitution in ErgA confers terbinafine resistance in Aspergillus fumigatus. Antimicrob Agents
Chemother. 2006;50:2533–6.
VetBooks.ir
328
J. D. Lemetayer and J. E. Sykes
139. Diekema DJ, Messer SA, Hollis RJ, Jones RN, Pfaller MA. Activities of caspofungin, itraconazole, posaconazole, ravuconazole, voriconazole, and amphotericin B against 448 recent
clinical isolates of filamentous fungi. J Clin Microbiol. 2003;41:3623–6.
140. Ndiritu CG, Enos LR. Adverse reactions to drugs in a veterinary hospital. J Am Vet Med
Assoc. 1977;171:335–9.
141. Wang A, Ding HZ, Liu YM, Gao Y, Zeng ZL. Single dose pharmacokinetics of terbinafine in
cats. J Feline Med Surg. 2012;14:540–4.
VetBooks.ir
Sporothrichosis
Hock Siew Han
Abstract
Sporothrix schenckii is currently recognized as a species complex consisting of
Sporothrix brasiliensis, Sporothrix schenckii sensu stricto, Sporothrix globosa,
and Sporothrix luriei. Due to divergent evolutionary process, each species possesses different virulence profiles, that allow it to thrive and persist in its niche.
Currently the disease in cats is primarily caused by S. brasiliensis, S. schenckii
sensu stricto and S. globosa, with cat fights and direct inoculation of the agent in
the skin as the main mode of disease transmission. Expression of putative virulence factors, such as adhesins, ergosterol peroxide, melanin, proteases, extracellular vesicles and thermotolerance, determines the clinical manifestation in the
feline patient, with thermotolerant S. brasiliensis exhibiting the highest pathogenicity, followed by S. schenckii sensu stricto, and S. globosa. Their ability to
produce biofilm is documented, but their clinical significance remains to be elucidated. Despite comprehensive descriptions of the pathogenicity of the agent
and of the disease, its prognosis remains guarded to poor, due to issues pertaining
to cost, protracted treatment course, zoonotic potential and low susceptibility of
some strains to antifungals.
Introduction
Sporothrix schenckii complex (also called S. schenckii sensu lato) causes a
chronic, granulomatous, cutaneous or subcutaneous infection, mainly occurring
in humans and cats. It has been recognised as an important cause of zoonotic
subcutaneous mycosis since its description by Dr. Benjamin Schenk in 1896 [1].
H. S. Han (*)
The Animal Clinic, Singapore
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_15
329
VetBooks.ir
330
H. S. Han
As a thermally dimorphic fungus, Sporothrix schenckii sensu lato exists as saprophyte in plant debris or decaying organic soil matter in its asexual filamentous
form (25–30 °C). With favourable temperature and environment (35–37 °C), it
phase transitions into its yeast form, and complete growth inhibition is achieved
at 40 °C, with no sexual reproduction observed to date [2]. This characteristic underpins the epidemiology of clinical sporotrichosis where historically, the
most common route of infection was reported to be the inoculation of conidia
into broken skin via contaminated soil during horticultural activities. It is only in
recent times that cats were perceived to be an important risk factor and disease
propagators [3–7].
Etiologic Agent
Sporothrix schenckii is currently recognized as a species complex consisting
of Sporothrix brasiliensis, Sporothrix schenckii sensu stricto, Sporothrix globosa,
and Sporothrix luriei (Clinical clade) based on DNA sequencing, with each species
having its own distinct virulence profiles and geographical distribution [8, 9]. S.
brasiliensis, S. s. sensu stricto and S. globosa, in order of virulence, are the main
species identified to cause pathology in cats [9]. S. brasiliensis, currently regionally restricted to Brazil, is characterised by its inherent thermotolerability which
is responsible for causing systemic spread. This species was identified as the main
cause of sporotrichosis epidemics in Rio de Janeiro and Sao Paolo, alongside S. s.
sensu stricto and S. globosa [10–12]. S. s. sensu stricto is the second most pathogenic species with a worldwide distribution, especially in tropical or subtropical
regions, with reports from the Americas, Africa, Australia and Asia. Zhou and colleagues demonstrated genetic diversity within this single species by subdividing S.
s. sensu stricto into clinical clade C (most commonly isolated from Americas and
Asia) and D (most commonly isolated from Americas and Africa), based on its
internal transcribed spacer (ITS) [13]. The recent identification of a single clonal
strain of S. s. sensu stricto clinical clade D from Malaysia (instead of the commonly isolated clinical clade C in Asia) suggests that this species is constantly
evolving, with the ability to undergo a process of selection and subsequent population expansion, depending on local environmental or host selection pressure [14,
15]. S. globosa is commonly identified as the species responsible for sporotrichosis
mainly in Asia and Europe, but is a rare cause in the Americas and Africa [11, 13,
16–20]. Exept S. pallida, Environmental clade associated sporothrix species such
as S.brunneoviolacea, S. lignivora, S. chilensis and S. mexicana (Sporothrix pallida complex) have not been reported to cause disease in the feline patient at the
time of writing [21]. These species are rare agents of sporotrichosis and normally
causes low virulence, opportunistic infections from traumatic inoculation of fungus from soil into host tissue. This is in contrast to sporothrix species within the
Clinical clade that is transmitted from animals.
Sporothrichosis
331
VetBooks.ir
Pathogenesis
Upon inoculation, the expression of putative virulence factors, such as adhesins, ergosterol peroxide, melanin, proteases, extracellular vesicles (EV) and
thermotolerance, determines the pathogenicity and clinical presentation of
sporotrichosis in the feline patient [22, 23]. The expression of adhesins and a
70 kDa glycoprotein (Gp70) on the cell wall mediates adhesion of the fungus
to fibronectin, type II collagen and laminin in the host [24]. Upon invasion,
the fungal cell wall composed of glucans, galactomannans, rhamnomannans,
chitin, glycoprotein, glycolipids and melanin provides the ability to survive
within host tissues and aids evasion from host innate immune response [25–
27]. Melanin production in both mycelial and yeast form shields against a
broad range of toxic insults. Melanin reduces susceptibility to antifungals and
enzymatic degradation, and confers protection against oxygen nitrogen free
radicals, macrophagic and neutrophilic phagocytosis [28]. The fungus readily
produces ergosterol peroxide and proteinases (Proteinase 1 and 2), which allow
it to evade phagocytosis and host immune response [29, 30]. EV (exosomes,
microvesicles and apoptotic bodies) are membranous compartments composed of lipid bilayers, released by all living cells to the extracellular medium,
that contain cargos of lipids (neutral glycolipids, sterols and phospholipids),
polysaccharides (glucuronoxylomannan, alpha-galactosyl epitopes), proteins
(lipases, proteases, urease, phosphatase) and nucleic acids (RNA) [31]. These
cargos represent virulence factors that contribute to drug resistance, facilitate
cell invasion and are eventually recognized by the innate immune system. EV
contribution to fungal virulence was described in Cryptococcus neoformans,
Histoplasma capsulatum, Paracoccidioides brasiliensis, Malassezia sympodialis, Candida albicans and, recently, also in Sporothrix brasiliensis [32–39].
Specifically, the EV cargos of Sporothrix brasiliensis, such as cell wall glucanase and heat shock proteins, were shown to increase phagocytosis but not
pathogen elimination, stimulate cytokine production (IL-12p40 and TNFα) and
favour the establishment of the fungus in the skin [38, 40, 41]. Current proteomic analyses revealed that 27% of EV proteins in S. brasiliensis and 35%
in S. schenckii remain to be characterized, including the identification of their
assigned biological process [38].
Thermotolerance, the ability of a fungus to grow or not at 37 °C, is another
important virulence factor that has been identified in Sporothrix spp. Isolates that
are able to grow at 35 °C but not at 37 °C in humans cause fixed cutaneous lesions,
but those that grow at 37 °C (a close approximation to human and animal core body
temperature) produce disseminated and extracutaneous lesions. Pathogenic thermotolerant species, such as S. brasiliensis have the ability to produce disseminated disease, compared to non-thermotolerant, less pathogenic species such as S. globosa.
S. s. sensu stricto displays variable thermotolerability [14].
VetBooks.ir
332
H. S. Han
The ability of Sporothrix schenckii complex to produce biofilm has recently been
documented, and an early report suggests that biofilm production alters the fungus
sensitivity to antifungals, however, the full extent of its clinical significance has yet
to be elucidated [42].
Both innate and adaptive immune responses play important roles in the prevention of disease progression. The first contact between fungal pathogen associated
molecular pattern (PAMPs) and host pattern recognition receptors (PPRs) is mediated by toll-like-receptors (TLR)-4 and TLR-2 [43, 44]. During the initiation of
infection, these receptors recognize lipid extracts from yeast cells that lead to an
increased production of tumour necrosis factor alpha (TNF-alpha), interleukin (IL)10 and nitric oxide (NO). While NO demonstrates antifungal activity in vitro, in vivo
it is associated with immunosuppression during the initial and the terminal stages
of the infection, due to its ability to increase apoptosis of immune cells [45]. The
role of NO in the infection was also documented in histoplasmosis by Histoplasma
capsulatum and paracoccidioidomycosis by Paracoccidioides brasiliensis [46, 47].
Yeast cells are also able to activate the antibody-dependent classical and alternative complement pathways [48, 49]. The main antigen recognized by antibodies is
a 70 kDa cell wall glycoprotein, named Gp70 [50]. This protein plays a crucial role
in fungal opsonisation, allowing macrophages to phagocytose and the production of
pro-inflammatory cytokines [51]. Nevertheless, the cornerstone for an effective fungal eradication is based on an effectively coordinated innate and adaptive immune
response (humoral and cell mediated) [52]. Recently, the nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome was shown to be critical to link the innate immune response to the adaptive
arm, contributing to effective protection against this infection by promoting the
production of pro-IL1β [53]. Fungal interaction with dendritic cells drives a mixed
Th1/Th17 immune response that activates macrophages, neutrophils and CD4+ T
cells, that release IFN-gamma, IL-12 and TNF-alpha that ultimately culminates in
the reduction of pathogen burden [54, 55].
Clinical Signs
Feline sporotrichosis occurs most commonly in young adult, free roaming intact
male cats and is associated with fighting, with no known breed predisposition [4].
In the human patient, clinical signs of sporotrichosis may be classified into 3 forms:
fixed cutaneous, lympho-cutaneous and disseminated forms, depending on the
pathogenicity of the fungal species and the status of host immunity (Fig. 1). Such
clear and distinct categorisation of clinical forms does not apply to cats and thus is
seldom used.
In cats, chronic non-healing lesions such as nodules, ulcers and crusts are commonly found on the head, especially at the bridge of the nose (Fig. 2), on the distal
limbs and tail base region (Fig. 3) and on the pinnae (Fig. 4). The majority of lesions
occur in cooler regions of the host body such as at the nasal passages and ear tips.
VetBooks.ir
Sporothrichosis
Fig. 1 A human patient
manifesting lymphocutaneous
sporotrichosis after being
bitten by a cat with
sporotrichosis (nodule at base
of thumb). Due to the lack of
thermotolerability of the
infectious agent, the lesion
did not progress beyond the
arm
Fig. 2 Classical
presentation of feline
sporotrichosis: chronic
non-healing wounds
affecting the bridge of the
nose
Fig. 3 Chronic nonhealing wounds affecting
the paws and the tail
333
334
b
VetBooks.ir
a
H. S. Han
Fig. 4 (a and b) Concave and convex aspects, respectively, of the pinna of a cat with sporotrichosis presenting numerous ulcerated nodules
If nasal passages are affected, extracutaneous signs such as sneezing, dyspnoea and
respiratory distress are commonly reported in tandem with cutaneous manifestations
[5]. Cutaneous screwworm myiasis as secondary infestation was recently reported
[56]. The fatal disseminated form of the disease is associated with S. brasiliensis
infection. Co-infection with either feline immunodeficiency virus (FIV) or feline
leukaemia virus (FeLV) has no significant effect on the clinical manifestations or on
the prognosis of the disease [57].
Diagnosis
A definitive diagnosis of feline sporotrichosis requires the isolation and identification of the agent in culture. The species identification can be obtained by
morphologic studies and physiologic phenotyping, as well as by polymerase
chain reaction targeting the calmodulin gene [5]. At 25–30°, the fungus exists in
its mycelial form and is seen as small and white or pale orange to orange-grey
colonies with no cottony aerial hyphae. Later, the colony becomes black, moist,
wrinkled, leathery or velvety with narrow white borders (Fig. 5). Some colonies
are however black from the onset. At 35–37°, yeast colonies are cream or tan,
smooth and yeast-like [2].
Cytologically, yeasts are found in abundance from cutaneous impression smears.
They are located intra- and extracellularly, in pleomorphic shapes, ranging from
VetBooks.ir
Sporothrichosis
335
Fig. 5 In its mature
mycelial form the fungi
becomes black, moist,
wrinkled, leathery or
velvety with narrow white
borders
Fig. 6 Cytologically, the
yeasts are found in
abundance intra- and
extracellularly in
pleomorphic shapes,
ranging from the classical
cigar-shaped to round or
oval, measuring 3–5 μm in
diameter with a thin, clear
halo around a pale blue
cytoplasm (Diff Quick,
1000×)
the classical cigar-shaped to round or oval bodies, measuring 3–5 μm in diameter
with a thin, clear halo around a pale-blue cytoplasm (Fig. 6) [58]. The sensitivity of
cytology to detect Sporothrix yeasts in the feline patient is estimated to range from
79% to 84.9% [59, 60].
On histology, a diffuse pyogranulomatous inflammation with large foci of necrosis is seen throughout the superficial and deep dermis, sometimes extending to the
subcutis. There are abundant round to cigar-shaped organisms, 3–10 μm in length
and 1–2 μm in diameter, seen both free and within macrophages. Commonly, organisms in cytoplasm of macrophages create large clear pockets full of yeast due to
poorly visualized yeast cell wall (Fig. 7) [61]. Periodic acid of Schiff (PAS) stain
may also be utilized to visualize yeasts as magenta stained organism on histological
preparation. Other diagnostic techniques such as serology (enzyme-linked immunosorbent assay, ELISA) and polymerase chain reaction (PCR) may also be used for
the diagnosis [62, 63].
VetBooks.ir
336
H. S. Han
Fig. 7 On histology there
are abundant round to
cigar-shaped organisms,
3–10 μm in length to
1–2 μm in diameter seen
both free and within
macrophages. Organisms
in cytoplasm of
macrophages create large
clear pockets full of yeasts
due to poorly visualized
yeast cell wall
Treatment
Treatment of feline sporotrichosis requires several months and must be continued
for at least 1 month beyond clinical cure. Luckily, despite a protracted treatment
course, it is current understanding that the fungus does not develop resistance during treatment [14].
Due to the high cost of treatment, high risk of therapeutic side effects and of
zoonosis and existence of low susceptibility strains, feline sporotrichosis carries a
guarded to poor prognosis. Currently, potassium iodide, azolic antifungals (ketoconazole, itraconazole), amphotericin B, terbinafine, local heat therapy, cryosurgery and surgical resection have all been documented as treatment options in the
feline patient. Potassium iodide has traditionally been the treatment of choice,
either in its saturated form (saturated salt of potassium iodide, SSKI) or in its
powder form re-­packaged into capsules. Dosages range from 10 to 20 mg/kg
every 24 hours [64, 65]. The powder form re-packaged into capsules is favoured
over SSKI for the feline patient, due to the latter’s tendency to cause hypersalivation. From a report of 48 cats receiving potassium iodide, 23 (47.9%) patients
achieved clinical cure with treatment failure in 18 cats (37.5%), two reported
deaths (4.2%) and treatment period averaging from 4 to 5 months. The most
commonly observed side effects were hyporexia, lethargy, weight loss, vomiting,
diarrhoea plus an increase in the liver enzyme alanine transaminase. No signs of
iodism (lacrimation, salivation, coughing, facial swelling, tachycardia) nor thyroid hormone abnormalities were observed in this study [64]. Due to its low cost,
potassium iodide is still often used either singularly or in conjunction with azole
antifungals to treat feline sporotrichosis [65].
VetBooks.ir
Sporothrichosis
337
Imidazoles such as ketoconazole and itraconazole currently represent the cornerstone therapy for feline sporotrichosis. Itraconazole is favoured over ketoconazole
as the latter is commonly associated with a higher rate of side effects, such a vomiting, hepatic dysfunction and altered cortisol metabolism. Itraconazole at 5–10 mg/
kg has been used successfully to treat feline sporotrichosis, with a maximum plasma
concentration of 0.7 ± 0.14 mg/L achieved after a 5 mg/kg oral dosing [66]. Based
on the updated Clinical and Laboratory Standards Institute (CLSI) reference method
for broth dilution antifungal susceptibility testing of filamentous fungi (document
M38-A2), the minimum inhibitory concentration (MIC) of antifungals against S.
brasiliensis, S. s sensu stricto and S. globosa is presented in Table 1 [14, 19, 20, 67,
68]. Itraconazole may be the treatment of choice but there are isolates with MIC
above 4 mg/L, the putative breakpoint for this antifungal agent. This variability
in MIC values may reflect the extensive divergent evolutionary process within the
Sporotrix complex, where each species developed its own repertoire of virulence
factors allowing thriving and persisting in its niche. Clinically, this is reflected by
the fact that some cases of feline sporotrichosis are refractory to treatment and thus
protocols based on higher dosages of itraconazole and/or its combination with other
antifungals have been explored to treat these refractory cases [65, 69]. Sporothrix
schenckii sensu lato generally displays low susceptibility towards fluconazole and
exhibits species-dependent susceptibility towards terbinafine and amphotericin B
(Table 1). Despite reports of successful treatment of human sporotrichosis with
terbinafine, results are still inconclusive for the feline patient [70, 71]. The recent
description of the protective effects of pyomelanin and eumelanin, synthesized by
S. brasiliensis and S. s. sensu stricto, against the antifungal terbinafine may partially
explain why in vitro results do not always correlate with in vivo responses when
patients are treated with this drug [72]. The administration of amphotericin B is
associated with toxicity, high cost and side effects, such as localized sterile abscess
formation from intralesional injections [5]. It is interesting to note that Sporothrix
spp. displays variable susceptibility towards antifungals rarely used in veterinary
medicine such as micafungin, 5-flucytosine and even posaconazole, highlighting
the importance of susceptibility testing [14, 20, 68]. Resolving granulomas are
visually and tactile-wise indistinguishable from normal adjacent healthy skin under
normal room lighting, and may be better visualized when held against a bright light
source (Fig. 8). Treatment should be continued for 1 month beyond the resolution
of all granulomas. Localized heat therapy is based on the fact that the fungus does
not grow at temperatures above 40 °C. This treatment modality, however, is associated with issues of practicality and perhaps welfare concerns in its application on
animals and has not been pursued as a feasible treatment option in the feline patient.
Cryosurgery, used in conjunction with itraconazole has been used successfully to
treat and cure 11 of 13 cats with sporotrichosis, with treatment lasting 3–16 months
and a median of 8 months [73]. Surgical resection is possible for localized singular
lesions but unpractical for generalized, disseminated forms.
S. brasiliensis
S. s. sensu stricto
S. globosa
40
9
61
5
32
23
Japan
Brazil
Iran
Brazil
Brazil
4
n
29
9
4
Malaysia
Iran
Origin
Japan subgroup I
Japan subgroup II
Brazil
Itraconazole
0.5–4
0.25–2
0.83
(0.06–16)
8
(1 -> 16)
1.3
(0.5–4)
0.5–1
0.42
(0.03–16)
0.76
(0.25–2)
2
0.36
(0.06–2)
Not tested
56.7
(16–128)
>128
57.7
(8–128)
>64
Fluconazole
>128
>128
53.8
(16–128)
>64
32 -> 64
>256
2
1.06
(0.03–2)
3.03
(1–8)
1.2
1.03
(0.2–4)
Amphotericin B
1–4
2–4
1
(0.2–4)
5.66
(4–8)
Not tested
Terbinafine
Not tested
Not tested
0.03
(0.01–0.06)
1.68
(1–2)
2.85
(1–8)
Not tested
0.05
(0.01–0.50)
0.38
(0.13–1)
0.1
0.06
(0.01–0.50)
[68]
[67]
[19]
[20]
[67]
[14]
[19]
References
[20]
[20]
[67]
Table 1 All results are expressed in mg/L and based on the Clinical and Laboratory Standards Institute (CLSI) reference method for broth dilution antifungal
susceptibility testing of filamentous fungi document M38-A2 (2008) in mycelial phase
VetBooks.ir
338
H. S. Han
Sporothrichosis
b
VetBooks.ir
a
339
Fig. 8 The author utilizes a bright light source to evaluate and ascertain cure. (a) A resolving
granulomatous reaction at the left ear tip, tactile and visually indistinguishable from adjacent normal tissue but is visualized with a bright light source. (b) Same patient after cure with complete
resolution of granuloma
Conclusion
The prognosis of feline sporotrichosis remains guarded to poor due to cost, protracted treatment course, risk of zoonosis and low susceptibility of some strains.
Despite the fact that antifungal susceptibility testing provides essential guidance for
the treatment, its lack of commercial availability and validated breakpoints remains
a stumbling block in the treatment of this disease. Unfortunately, the current repertoire of veterinary antifungals classes are inadequate to address the issue of fungal
low fungal susceptibility.
References
1. Schenck BR. On refractory subcutaneous abscess caused by a fungus possibly related to the
Sporotricha. Bull Johns Hopkins Hosp. 1898;9:286–90.
2. Larone DH. Identification of fungi in culture. In: Medically important fungi: a guide to identification. 5th ed. Washington, DC: ASM Press; 2011. p. 166–7.
3. Schubach A, Schubach TM, Barros MB, Wanke B. Cat-transmitted sporotrichosis, Rio de
Janeiro, Brazil. Emerg Infect Dis. 2005;11(1):1952–4.
4. Rodrigues AM, de Hoog GS, de Camargo ZP. Sporothrix Species Causing Outbreaks in Animals
and Humans Driven by Animal–Animal Transmission. PLoSPathog. 2016;12:e1005638.
https://doi.org/10.1371/journal.ppat.100.
VetBooks.ir
340
H. S. Han
5. Gremião ID, Menezes RC, Schubach TM, Figueiredo AB, Cavalcanti MC, Pereira SA. Feline
sporotrichosis: epidemiological and clinical aspects. Med Mycol. 2015;53(1):15–21.
6. Gremião IDF, Miranda LHM, Reis EG, Rodrigues AM, Pereira AS. Zoonotic epidemic of
sporotrichosis: cat to human transmission. PLoS Pathog. 2017;13(1):1–7.
7. Tang MM, Tang JJ, Gill P, Chang CC, Baba R. Cutaneous sporotrichosis: a six-year review of
19 cases in a tertiary referral center in Malaysia. Int J Dermatol. 2012;51:702–8.
8. Marimon R, Cano J, Gene J, Sutton DA, Kawasaki M, Guarro J. Sporothrix brasiliensis,
S. globosa, and S. mexicana, three new Sporothrix species of clinical interest. J Clin Microbiol.
2007;45:3198-206.
9. Arrillaga-Moncrieff CJ, Mayayo E, Marimon R, Marine M, Gene J, et al. Different virulence
levels of the species of Sporothrix in a murine model. Clin Microbiol Infect. 2009;15:651–5.
10. Rodrigues AM, de Melo Teixeira M, de Hoog GS, TMP S, Pereira SA, Fernandes GF, et al.
Phylogenetic analysis reveals a high prevalence of Sporothrix brasiliensis in feline sporotrichosis outbreaks. PLoS Negl Trop Dis. 2013;7(6):e2281.
11. Oliveira MME, Almeida-Paes R, Muniz MM, Barros MBL, Gutierrez-Galhardo MC, Zancope-­
Oliveira RM. Sporotrichosis caused by Sporothrix globosa in Rio de Janeiro, Brazil: case
report. Mycopathologia. 2010;169:359–63.
12. Oliveira MME, Almeida-Paes R, Muniz MM, Gutierrez-Galhardo MC, Zancope-Oliveira
RM. Phenotypic and molecular identification of Sporothrix isolates from an epidemic area of
sporotrichosis in Brazil. Mycopathologia. 2011;172(4):257–67.
13. Zhou X, Rodrigues A, Feng P, Hoog GS. Global ITS diversity in the Sporothrix schenckii
complex. Fungal Divers. 2013:1–13.
14. Han HS, Kano R, Chen C, Noli C. Comparisons of two in vitro antifungal sensitivity tests
and monitoring during therapy of Sporothrix schenckii sensu stricto in Malaysian cats. Vet
Dermatol. 2017;28:156–e32.
15. Kano R, Okubo M, Siew HH, Kamata H, Hasegawa A. Molecular typing of Sporothrix
schenckii isolates from cats in Malaysia. Mycoses. 2015;58:220–4.
16. Watanabe M, Hayama K, Fujita H, Yagoshi M, Yarita K, Kamei K, et al. A case of Sporotrichosis
caused by Sporothrix globosa in Japan. Ann Dermatol. 2016;28:251–2.
17. Yu X, Wan Z, Zhang Z, Li F, Li R, Liu X. Phenotypic and molecular identification of Sporothrix
isolates of clinical origin in Northeast China. Mycopathologia. 2013;176:67–74.
18. Madrid H, Cano J, Gene J, Bonifaz A, Toriello C, Guarro J. Sporothrix globosa, a pathogenic
fungus with widespread geographical distribution. Rev Iberoam Micol. 2009;26(3):218–22.
19. Mahmoudi S, Zaini F, Kordbacheh P, Safara M, Heidari M. Sporothrix schenckii complex in
Iran: molecular identification and antifungal susceptibility. Med Mycol. 2016;54:593–9.
20. Suzuki R, Yikelamu A, Tanaka R, Igawa K, Yokodeki H, Yaguchi T. Studies in phylogeny,
development of rapid identification methods, antifungal susceptibility and growth rates of
clinical strains of Sporothrix schenckii Complex in Japan. Med Mycol J. 2016;57E:E47–57.
21. Thomson J, Trott DJ, Malik R, Galgut B, McAllister MM, Nimmo J et al. An atypical cause of
sporotrichosis in a cat. Med Mycol Case Reports. 2019;23:72-6.
22. Barros MB, Paes RA, Schubach AO. Sporothrix schenckii and Sporotrichosis. Clin Microbiol
Rev. 2011;24:633–54.
23. Rossato L, Moreno F, Jamalian A, Stielow B, Almeida R, de Hoog S, et al. Proteins potentially
involved in immune evasion strategies in Sporothrix brasiliensis elucidated by high resolution
mass spectrometry. mSphere. 2018;13:e00514–7.
24. Teixeira PA, de Castro RA, Nascimento RC, Tronchin G, Torres AP, Lazéra M, et al. Cell surface expression of adhesins for fibronectin correlates with virulence in Sporothrix schenckii.
Microbiology. 2009;155:3730–8.
25. López-Esparza A, Álvarez-Vargas A, Mora-Montes HM, Hernández-Cervantes A, Del Carmen
C-CM, Flores-Carreón A. Isolation of Sporothrix schenckii GDA1 and functional characterization of the encoded guanosine diphosphatase activity. Arch Microbiol. 2013;195:499–506.
26. Morris-Jones R, Youngchim S, Gomez BL, Aisen P, Hay RJ, Nosanchuk JD, et al. Synthesis of
melanin-like pigments by Sporothrix schenckii in vitro and during mammalian infection. Infect
Immun. 2003;71:4026–33.
VetBooks.ir
Sporothrichosis
341
27. Teixeira PA, De Castro RA, Ferreira FR, Cunha MM, Torres AP, Penha CV, et al. L-DOPA
accessibility in culture medium increases melanin expression and virulence of Sporothrix
schenckii yeast cells. Med Mycol. 2010;48:687–95.
28. Nosanchuk JD, Casadevall A. Impact of melanin on microbial virulence and clinical resistance
to antimicrobial compounds. Antimicrob Agents Chemother. 2006;50:3519–28.
29. Sgarbi DB, da Silva AJ, Carlos IZ, Silva CL, Angluster J, Alviano CS. Isolation of ergosterol peroxide and its reversion to ergosterol in the pathogenic fungus Sporothrix schenckii.
Mycopathologia. 1997;139:9–14.
30. Lei PC, Yoshiike T, Ogawa H. Effects of proteinase inhibitors on cutaneous lesion of Sporothrix
schenckii inoculated hairless mice. Mycopathologia. 1993;123:81–5.
31. Joffe LS, Nimrichter L, Rodrigues ML, Del Poeta M. Potential roles of fungal extracellular
vesicles during infection. mSphere. 2016;1:e00099–16.
32. Rodrigues ML, Nimrichter L, Oliveira DL, Frases S, Miranda K, Zaragoza O, et al. Vesicular
polysaccharide export in Cryptococcus neoformans is a eukaryotic solution to the problem of
fungal trans-cell wall transport. Eukaryot Cell. 2007;6:48–59.
33. Rodrigues ML, Nimrichter L, Oliveira DL, Nosanchuk JD, Casadevall A. Vesicular trans-cell
wall transport in fungi: a mechanism for the delivery of virulence-associated macromolecules?
Lipid Insights. 2008;2:27–40.
34. Albuquerque PC, Nakayasu ES, Rodrigues ML, Frases S, Casadevall A, Zancope-Oliveira
RM, et al. Vesicular transport in Histoplasma capsulatum: an effective mechanism for trans-­
cell wall transfer of proteins and lipids in ascomycetes. Cell Microbiol. 2008;10:1695–710.
35. Vallejo MC, Matsuo AL, Ganiko L, Medeiros LC, Miranda K, Silva LS, et al. The pathogenic fungus Paracoccidioides brasiliensis exports extracellular vesicles containing highly
immunogenic-­galactosyl epitopes. Eukaryot Cell. 2011;10:343–51.
36. Vargas G, Rocha JD, Oliveira DL, Albuquerque PC, Frases S, Santos SS, et al. Compositional
and immunobiological analyses of extracellular vesicles released by Candida albicans. Cell
Microbiol. 2015;17:389–407.
37. Rayner S, Bruhn S, Vallhov H, Anderson A, Billmyre RB, Scheynius A. Identification of small
RNAs in extracellular vesicles from the commensal yeast Malassezia sympodialis. Sci Rep.
2017;7:39742.
38. Ikeda MAK, de Almeida JRF, Jannuzzi GP, Cronemberger-Andrade A, Torrecilhas ACT,
Moretti NS, et al. Extracellular vesicles from Sporothrix brasiliensis are an important virulence factor that induce an increase in fungal burden in experimental sporotrichosis. Front
Microbiol. 2018;9:2286.
39. Huang SH, Wu CH, Chang YC, Kwon-Chung KJ, Brown RJ, Jong A. Cryptococcus
neoformans-­derived microvesicles enhance the pathogenesis of fungal brain infection. PLoS
One. 2012;7:e48570.
40. Rossato L, Moreno F, Jamalian A, Stielow B, Almeida R, de Hoog S, et al. Proteins potentially
involved in immune evasion strategies in Sporothrix brasiliensis elucidated by ultra-high-­
resolution mass spectrometry. mSphere. 2018;3:e00514–7.
41. Nimrichter L, de Souza MM, Del Poeta M, Nosanchuk JD, Joffe L, Tavares PM, Rodrigues
ML. Extracellular vesicle-associated transitory cell wall components and their impact on the
interaction of fungi with host cells. Front Microbiol. 2016;7:1034.
42. Brilhante RSN, de Aguiar FRM, da Silva MLQ, de Oliveira JS, de Camargo ZP, Rodgrigues
AM, et al. Antifungal susceptibility of Sporothrix schenckii complex biofilms. Med Mycol.
2018;56:297–306.
43. Carlos IZ, Sassá MF, da Graca Sgarbi DB, MCP P, DCG M. Current research on the immune
response to experimental sporotrichosis. Mycopathologia. 2009;168:1–10.
44. Negrini Tde C, Ferreira LS, Alegranci P, Arthur RA, Sundfeld PP, Maia DC, et al. Role of
TLR-2 and fungal surface antigen on innate immune response against Sporothrix schenckii.
Immuno Invest. 2013;42:36–48.
45. Fernandes KS, Neto EH, Brito MM, Silva JS, Cunha FQ, Barja-Fidalgo C. Detrimental
role of endogenous nitric oxide in host defense againsts Sporothrix schenckii. Immunology.
2008;123:469–79.
VetBooks.ir
342
H. S. Han
46. Brummer E, Division DA. Antifungal mechanism of activated murine bronchoalveolar or peritoneal macrophages for Histoplasma capsulatum. Clin Exp Immunol. 1995;102:65–70.
47. Bocca L, Hayashi EE, Pinheiro G, Furlanetto B, Campanelli P, Cunha FQ, et al. Treatment of
Paracoccidioides brasiliensis-infected mice with a nitric oxide inhibitor prevents the failure of
cell-mediated immune response. J Immunol. 1998;161:3056–63.
48. Torinuki W, Tagami H. Complement activation by Sporothrix schenckii. Arch Dermatol Res.
1985;277:332–3.
49. de Lima FD, Nascimento RC, Ferreira KS, Almeida SR. Antibodies against Sporothrix
schenckii enhance TNF-alpha production and killing by macrophages. Scand J Immunol.
2012;75:142–6.
50. Ruiz-Baca E, Toreillo C, Perez-Torres A, Sabanero-López M, Villagómez-Castro JC, López-­
Romero E. Isolation and some properties of a glycoprotein of 70 kDa (Gp70) from the cell wall
of Sporothrix shcenckii cell wall. Mem Inst Oswaldo Cruz. 2009;47:185–96.
51. Maia DC, Sassá MF, Placeres MC, Carlos IZ. Influence of Th1/Th2 cytokines and nitric oxide in
murine systemic infection induced by Sporothrix schenckii. Mycopathologia. 2006;161:11–9.
52. Plouffe JF, Silva J, Fekety R, Reinhalter E, Browne R. Cell-mediated immune responses III
sporotrichosis. J Infect Dis. 1979;139:152–7.
53. Goncalves AC, Ferreira LS, Manente FA, de Faria CMQG, Polesi MC, de Andrade CR, et al.
The NLRP3 inflammasome contributes to host protection during Sporothrix schenckii infection. Immunology. 2017;151:154–66.
54. Tachibana T, Matsuyama T, Mitsuyama M. Involvement of CD4+ T cells and macrophages
in acquired protection against infection with Sporothrix schenckii in mice. Med Mycol.
1999;37:397–404.
55. Flores-García A, Velarde-Félix JS, Garibaldi-Becerra V, Rangel-Villalobos H, Torres-Bugarín
O, Zepeda-Carrillo EA, et al. Recombinant murine IL-12 promotes a protective TH1/cellular
response in Mongolian gerbils infected with Sporothrix schenckii. J Chemother. 2015;27:87–93.
56. Han HS, Toh PY, Yoong HB, Loh HM, Tan LL, Ng YY. Canine and feline cutaneous screw-­
worm myiasis in Malaysia: clinical aspects in 76 cases. Vet Dermatol. 2018;29:442–e148.
57. Schubach TM, Schubach A, Okamoto T, Barros MB, Figueiredo FB, Cuzzi T, et al. Evaluation
of an epidemic of sporotrichosis in cats: 347 cases (1998–2001). J Am Vet Med Assoc.
2004;224(10):623–9.
58. Raskin RE, Meyer DJ. Skin and subcutaneous tissue. In: Canine and feline cytology. 2nd ed.
St. Louis: Saunders Elsevier; 2010. p. 41–4.
59. Pereira SA, Menezes RC, Gremião ID, Silva JN, Honse Cde O, Figueiredo FB, et al. Sensitivity
of cytopathological examination in the diagnosis of feline sporotrichosis. J Feline Med Surg.
2011;13:220–3.
60. Jessica N, Sonia RL, Rodrigo C, Isabella DF, Tânia MP, Jeferson C, et al. Diagnostic accuracy assessment of cytopathological examination of feline sporotrichosis. Med Mycol.
2015;53(8):880–4.
61. Gross TL, Ihrke PJ, Walder EJ, et al. Infectious nodular and diffuse granulomatous and pyogranulomatous diseases of the dermis. In: Skin disease of the dog and cat. 2nd ed. Oxford:
Blackwell Science; 2005. p. 298–301.
62. Fernandes GF, Lopes-Bezerra LM, Bernardes-Engemann AR, Schubach TM, Dias MA, Pereira
SA, et al. Serodiagnosis of sporotrichosis infection in cats by enzyme-linked immunosorbent
assay using a specific antigen, SsCBF, and crude exoantigens. Vet Microbiol. 2011;147:445–9.
63. Kano R, Watanabe K, Murakami M, Yanai T, Hasegawa A. Molecular diagnosis of feline sporotrichosis. Vet Rec. 2005;156:484–5.
64. Reis EG, Gremião ID, Kitada AA, Rocha RF, Castro VP, Barros ML, et al. Potassium iodide
capsule treatment of feline sporotrichosis. J Fel Med Surg. 2012;14:399–404.
65. Reis ÉG, Schubach TM, Pereira SA, Silva JN, Carvalho BW, Quintana MB, et al. Association
of itraconazole and potassium iodide in the treatment of feline sporotrichosis: a prospective
study. Med Mycol. 2016;54:684–90.
66. Liang C, Shan Q, Zhang J, Li W, Zhang X, Wang J, et al. Pharmacokinetics and bioavailability
of itraconazole oral solution in cats. J Fel Med Surg. 2016;18:310–4.
VetBooks.ir
Sporothrichosis
343
67. Ottonelli Stopiglia CD, Magagnin CM, Castrillón MR, Mendes SD, Heidrich D, Valente P,
et al. Antifungal susceptibility and identification of Sporothrix schenckii complex isolated in
Brazil. Med Mycol. 2014;52:56–64.
68. Borba-Santos LP, Rodrigues AM, Gagini TB, Fernandes GF, Castro R, de Camargo ZP, et al.
Susceptibility of Sporothrix brasiliensis isolates to amphotericin B, azoles, and terbinafine.
Med Mycol. 2015;53:178–88.
69. Han HS. The current status of feline sporotrichosis in Malaysia. Med Mycol J.
2017;58E:E107–13.
70. Francesconi G, Valle AC, Passos S, Reis R, Galhardo MC. Terbinafine (250mg/day): an
effective and safe treatment of cutaneous sporotrichosis. J Eur Acad Dermatol Venereol.
2009;23:1273–6.
71. Vettorato R, Heidrich D, Fraga F, Ribeiro AC, Pagani DM, Timotheo C, et al. Sporotrichosis
by Sporothrix schenckii sensu stricto with itraconazole resistance and terbinafine sensitivity
observed in vitro and in vivo: case report. Med Mycol Case Reports. 2018;19:18–20.
72. Almeida-Paes R, Figueiredo-Carvalho MHG, Brito-Santos F, Almeida-Silva F, Oliveira MME,
Zancopé-Oliveira RM. Melanins protect Sporothrix brasiliensis and Sporothrix schenckii from
the antifungal effects of terbinafine. PLoS One. 2016;11:e0152796. https://doi.org/10.1371/
journal.pone.0152796.
73. De Souza CP, Lucas R, Ramadinha RH, Pires TB. Cryosurgery in association with itraconazole for the treatment of feline sporotrichosis. J Feline Med Surg. 2016;18:137–43.
VetBooks.ir
Malassezia
Michelle L. Piccione and Karen A. Moriello
Abstract
Malassezia dermatitis/overgrowth is a superficial fungal (yeast) skin disease of
cats. It has most often been reported in association with underlying hypersensitivity skin diseases, metabolic diseases, neoplasia and paraneoplastic syndromes. Common clinical signs include dark waxy debris associated with otitis
externa, scaling, black waxy nail bed debris (paronychia), variable pruritus,
erythema, and exudative dermatitis especially when complicated by bacterial
pyoderma. The disease is most commonly diagnosed by cytological examination of the skin. Malassezia pachydermatis is the primary species isolated from
cats; however, other lipid-dependent species can be isolated. Itraconazole is the
treatment of choice along with topical antifungal shampoo therapy or leave-on
antifungal products. Recurrent Malassezia dermatitis is a clinical sign of an
underlying trigger, most of which are not life threatening. In cats with severe
widespread disease, especially those with erythema, alopecia and/or marked
scaling, Malassezia species overgrowth could be a clinical sign of systemic
disease warranting a thorough systemic evaluation.
Introduction
Malassezia are yeast organisms and are part of the normal cutaneous microflora of
humans and animals, including cats [1]. At the time of writing, at least 16 different
human and animal species have been isolated. A review of the literature revealed a
wide range of species isolated from cats but molecular diagnostics are reclassifying
M. L. Piccione (*) · K. A. Moriello
School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
e-mail: mpiccione@wisc.edu; karen.moriello@wisc.edu
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_16
345
VetBooks.ir
346
M. L. Piccione and K. A. Moriello
some of these species [2]. Several recent studies reconfirmed that the most commonly isolated species from cats are M. pachydermatis, M. furfur, M. nana, and M.
sympodialis [3–6].
For the purposes of this chapter, the term “Malassezia dermatitis” and “Malassezia
overgrowth” are synonymous and the former will be used for simplicity. Malassezia
spp. dermatitis is increasingly being recognized as a complicating factor in many
feline skin diseases, often in association with bacterial overgrowth. The goal of this
chapter is to review the scientific literature available on Malassezia dermatitis in
cats and summarize the key aspects of clinical signs, diagnosis, and treatment.
Etiology and Pathogenesis of Feline Malassezia
Biological Characteristics
The genus Malassezia are lipophilic yeast, are part of the cutaneous microflora
of warm-blooded animals and tend to colonize skin rich in sebaceous glands.
Malassezia belong to the basidiomycetous yeasts. They are characterized by a
multilayered cell wall and reproduce by unilateral budding [7]. Bottle-shaped
yeast may be globose, ovoid, or cylindrical. Buds form on a narrow or wide base
[7]. Currently, the genus Malassezia includes 16 species, of which 15 are lipiddependent and are most frequently recovered from humans, ruminants, and horses
(Malassezia furfur, M. globosa, M. obtusa, M. restricta, M. slooffiae, M. sympodialis, M. dermatis, M. nana, M. japonica, M. yamatoensis, M. equina, M. caprae
and M. cuniculi, M. brasiliensis, M. psittaci) [8]. The only non-lipid-dependent
species, M. pachydermatis, is commonly recovered from cats and dogs [8]. With
the exception of M. pachydermatis, the lipophilic yeast requires supplementation
of long chain fatty acids in culture medium; utilizing the lipids is a source of carbon for survival [8].
Pathogenesis
M. pachydermatis is considered a nonpathogenic, commensal organism that can
become an opportunistic pathogen when the environmental factors are appropriate and/or the host’s defense mechanism fails. Factors involved in maintaining the
balance of skin microflora include temperature, hydration, chemical constituents
(sweat, sebum, and saliva) and pH [9]. When these factors are altered, Malassezia
can overgrow and act as a pathogen on the skin of cats, inducing an inflammatory
response. M. pachydermatis has been shown to adhere to human keratinocyte cells
and keratinocytes respond by releasing pro-inflammatory mediators as a defense
mechanism [10]. Humoral and cell-mediated responses to Malassezia have been
documented and anything that interferes with or blunts these responses can result
in overgrowth [11].
Malassezia
347
VetBooks.ir
Prevalence
There are many studies investigating the carriage of Malassezia in healthy cats, cats
with skin disease, in specific cat breeds, on various skin sites and in association with
other diseases. It is important to keep in mind that reported studies used different
methodologies (i.e., culture, cytology, combination of culture and cytology) making
direct comparisons difficult.
In one study comparing 10 domestic short-haired (DSH) cats with no history
of skin disease (controls) and 32 Sphynx cats, Malassezia was not isolated from
the skin of control cats [12]. In Sphynx cats, it was isolated from 26 of 32 cats
of which 5 were reported to have greasy skin (Fig. 1). There were 73 isolates of
Malassezia, of which 69 were M. pachydermatis. Interestingly, Malassezia was not
isolated from the ears of any of the 42 cats. In another study, carriage was compared
between several groups of cats: 10 normal DSH, 33 Cornish Rex cats (5 normal, 28
with seborrheic skin disease), and 30 Devon Rex cats (21 normal and 9 with seborrheic skin disease) [13]. Malassezia was isolated in 5 of 10 normal cats, 5 of 15
Cornish Rex cats, and 27 of 30 Devon Rex cats. When normal and diseased cat data
was pooled, M. pachydermatis was isolated from 70% of cats with seborrheic skin
disease and only 17% of cats with normal skin. In this study, 121 of 141 isolates
were M. pachydermatis.
The prevalence of Malassezia in the ear canal of cats is an area of interest, since
otitis is a common problem in cats (Chapter, Otitis) (Fig. 2). In one study, Malassezia
species were isolated from the ear canal of 63 of 99 (63.6%) cats with otitis and 12
of 52 (23%) normal cats [14]. In this study, M. pachydermatis, M. globosa, and M.
furfur were the most common isolates. In another study, Malassezia was isolated
from 9 of 17 cats with otitis externa and in 16 of 51 cats without otitis [15]. Again,
M. pachydermatis, M. globosa, and M. furfur were the most common isolates. In
Fig. 1 Sphynx cat with
seborrheic dermatitis and
yeast overgrowth. This cat
was very pruritic and yeast
organisms were found on
cytology from the papular
eruption shown. The cat
was diagnosed with
environmental allergies
VetBooks.ir
348
M. L. Piccione and K. A. Moriello
Fig. 2 Ear of cat with
yeast otitis. This is the
pinna of a cat with
hypersensitivity dermatitis.
The cat has extremely
pruritic ears and it
responded well to topical
steroid treatment
Fig. 3 Nail fold of a cat
with Malassezia dermatitis.
The owner reported that
this cat licked and chewed
at the paws and nail fold
area. The lesions resolved
with topical and systemic
antifungal treatment. The
cat had concurrent diabetes
mellitus
yet another study involving normal and affected cats, Malassezia was isolated in 7
of 25 and 15 of 20 cats [16]. Again, M. pachydermatis and M. sympodialis were the
most common isolates.
Another interesting site that has been investigated in cats is the nail fold (Fig. 3).
In one study, yeast were isolated from the claw fold of 26 of 29 Devon Rex cats [17].
In another study, Malassezia was found in 28 of 46 nail fold samplings from cats
[3]. Yeast were found in all 15 Devon Rex cats, 10 DSH cats, and 3 Persian cats.
Malassezia are also commonly found in the nail folds of Sphynx cats [4].
The facial fold of Persian cats has also been investigated for Malassezia dermatitis [18]. This breed is well known to have facial fold dermatitis which is often
idiopathic in origin. In one clinical case series, 13 Persian cats with idiopathic facial
fold dermatitis were investigated and Malassezia dermatitis was found in 6 of 13
cats. There was an incomplete response to treatment, suggesting Malassezia was
more of a complicating factor than a cause.
VetBooks.ir
Malassezia
349
Prevalence studies show some common trends. First, Malassezia can be found
on healthy cats but it is not common. Carriage is more common in cat breeds with
genetically associated follicular dysplasia (Devon Rex, Cornish Rex, and Sphynx
cat breeds). Interestingly, although Cornish Rex and Devon Rex cats share similar coat characteristics, the frequency and population of Malassezia isolated are
different. The degree of colonization may be associated with the Devon Rex cat’s
predisposition to development of seborrheic dermatitis. Malassezia can be isolated
from cats with and without otitis externa and from the nail folds of cats, particularly
those with seborrheic or allergic skin disease. Finally, M. pachydermatis is the most
common Malassezia isolate from cats.
Malassezia and Concurrent Diseases
Malassezia overgrowth/dermatitis is a common complication of skin diseases in
other species and a similar picture is starting to emerge in cats.
Hypersensitivity skin disease is common in cats and the role of Malassezia dermatitis is increasingly being recognized (Fig. 4). In one study of 18 cats with hypersensitivity dermatitis, Malassezia dermatitis was found in all cats [19]. Sixteen cats
showed marked reduction in pruritus after treatment. This suggests Malassezia
overgrowth may be a contributing factor in some cats with hypersensitivity dermatitis. Not all cats with hypersensitivity skin disease have Malassezia dermatitis. A
molecular study on fungal microbiota of allergic cats (n = 8) found Malassezia in
only 21% of 54 samples from the 8 allergic cats [20].
A study of aural microflora in healthy cats (n = 20) compared with allergic cats
(n = 15) and cats with systemic disease (n = 15) found that Malassezia colonization
was more common in cats with hypersensitivity dermatitis and systemically ill cats
compared to healthy cats [21]. In another study, Malassezia was more commonly
isolated from retroviral positive cats than retroviral negative cats [22]. Although the
cats were healthy, possibly retroviral infections interfered with the innate immune
response. When the frequency of isolation of Malassezia on the skin of cats with
Fig. 4 This cat had
hypersensitivity dermatitis
and recurrent areas of
eosinophilic dermatitis.
Cytology revealed
concurrent bacterial and
Malassezia dermatitis.
Lesions resolved with
concurrent antibacterial
and antifungal therapy
VetBooks.ir
350
M. L. Piccione and K. A. Moriello
diabetes mellitus (n = 16), hyperthyroidism (n = 20, and neoplasia were compared
(n = 8) to normal cats (n = 10), no difference was found [23].
There is increasing evidence that Malassezia dermatitis may be associated with
paraneoplastic alopecia and/or be a cutaneous sign of systemic disease. In the above
study, Malassezia was isolated from 9 sites in one cat with feline paraneoplastic
syndrome and pancreatic adenocarcinoma [23]. In another case report, marked exfoliative dermatitis and yeast overgrowth was found in a cat with thymoma (Figs. 5
and 6) and, interestingly, there was complete resolution of clinical signs after complete surgical tumor resection [24]. One case report described a 13-year-old DSH
cat with a history of progressively worsening paraneoplastic alopecia along with
Malassezia overgrowth. Post-mortem results revealed a pancreatic adenocarcinoma
with hepatic metastases [25]. In a retrospective study evaluating skin biopsy specimens from cats, 15 specimens contained large numbers of Malassezia organisms
Fig. 5 This is a 13-yearold cat that presented with
marked exfoliative
dermatitis with severe
Malassezia dermatitis
found on cytology. The cat
was systemically ill.
Imaging revealed a
thymoma
Fig. 6 This is a close-up
view of the marked
exfoliation on the skin of
the cat in Fig. 5. Note
marked erythema and large
sheets of shed
keratinocytes. This
appearance of scales is
highly suggestive of feline
exfoliative dermatitis due
to an underlying medical
problem which may or
may not be associated with
a thymoma
VetBooks.ir
Malassezia
351
in the epidermis or follicular infundibulum [26]. When clinical data was evaluated,
11 of 15 cats had acute onset of multifocal to generalized skin lesions. All 10 cats
were euthanized and one died of metastatic carcinoma of the liver 2 months after
the onset of clinical signs.
Clinical Signs
There are no pathognomonic clinical signs for Malassezia dermatitis in cats.
Clinical signs reported and/or commonly noted by the authors are summarized in
Box 1 and chapter images. Concurrent bacterial overgrowth is common. Scaling
and an unkempt coat (Fig. 8) is a common finding and often Malassezia is found on
cytology. Many cats with Malassezia dermatitis due to poor grooming will respond
to coat hygiene and topical therapy alone. Nail bed involvement may vary in clinical
appearance, it is usually brown black (Fig. 3) and may appear as marked seborrheic
accumulations.
Box 1: Clinical Signs of Malassezia Dermatitis/Overgrowth
•
•
•
•
•
•
•
•
•
•
•
•
•
Lesions can be generalized or localized
Pruritus varies from none to marked
Erythema
Diffuse seborrhea that can be dry and/or oily
Increased scaling
Hairs pierced by scales and follicular casts
Traumatic alopecia
Hyperpigmentation characterized by brown waxy exudate
Follicular plugging on ventral abdomen, especially around nipples
Brown to reddish brown discoloration of nails
Waxy debris under the nail folds
Brown waxy debris adherent to lip folds
Chin acne
Malassezia otitis
•
•
•
•
Increased ceruminous debris
Erythema of ear canal
Swelling and narrowing of canal
Pruritus of pinna and/or canal
VetBooks.ir
352
M. L. Piccione and K. A. Moriello
Fig. 7 Cat with
Malassezia and bacterial
overgrowth. Note the
erythema, eruptions, and
scaling
Fig. 8 Malassezia
dermatitis in a cat with an
unkempt coat
Diagnosis
The diagnosis of Malassezia dermatitis is based the identification of organism in
light of a compatible/plausible history and clinical signs and a good response to
antifungal treatment.
Cytology
Cytological examination of the skin is the single best technique for investigating whether or not Malassezia are present. There are no cytological criteria for
VetBooks.ir
Malassezia
353
determining the “normal number” of Malassezia organisms present on the skin of
cats. The presence or absence of organisms can only be interpreted in light of the
cat’s clinical signs. The organisms are much larger than bacteria and can vary in size
from 2–4 μm by 3 to 7 μm.
Skin cytology samples can be obtained using a clear acetate tape which allows
for sampling in difficult areas, e.g., facial, interdigital, or nail folds. A clear piece
of tape is pressed to the skin, the tape is then stained using in-house cytology
stains (e.g., Diff Quik). It is important to avoid the fixative step and to stain the
tape by holding with forceps, tweezers, or the authors’ favorite tool, household
clothes pins. Affixing unstained tape to a glass slide and then staining the slide
results in poor staining and increased artifacts, and should be avoided. To make
a proper preparation, put a drop of immersion oil on a glass slide, then mount
a thoroughly dry stained piece of tape over the oil, and then examine it microscopically. For oil immersion, (recommended), a drop of immersion oil can be
placed directly over the tape. Glass slide samples are the optimum tool to use
when sampling the skin. To obtain the best possible sample, place the glass slide
over the target area, gently lift the skin and squeeze the skin between two fingers.
This will markedly increase the cellularity of the sample. Ears are best sampled
with a cotton tip applicator. Nail beds are best sampled by gently scraping debris
from under the claw fold using a skin scraping spatula, not a scalpel blade, and
then smearing it onto a glass slide. In all cases, it is important to NOT heat fix
slides as this will cause artifacts and/or damage other cells on the slide. It has
been shown that increasing the number of dips in solution II (basophilic, blue)
is all that is needed to improve visualization of yeast organisms [27, 28]. The
authors routinely examine slides at 4× to find a cellular area, 10× and then 100×.
Malassezia organisms are variable in size and may be seen easily on the slide or
adherent to skin keratinocytes (Figs. 9 and 10).
Fig. 9 Note the large
number of Malassezia
organisms in this ear
cytology. There are
peanut- and ovoid-shaped
organisms. Some of the
organisms have not stained
as deeply basophilic and
this is common in samples
from ear cytology
VetBooks.ir
354
M. L. Piccione and K. A. Moriello
Fig. 10 Note the large
number of Malassezia
organisms adherent to skin
cells. This sample was
obtained from a cat with
exfoliative dermatitis. Note
the concurrent bacteria in
this sample
Fungal Culture
It is rare to need to culture this organism for diagnosis in clinical cases. If there
is a need, e.g., suspected antifungal resistance, for research or if there is need to
identify the species, there are two important things to remember. First, if using a
culture swab to culture the skin, moisten the swab with the transport medium and
aggressively rub the swab over a large area of the skin while rotating the head of
the swab 360°. In the authors’ experience, dry swab cultures of small areas are
inadequate. If available, contact plate cultures are preferable. Secondly, it is common to isolate several different species from cats. It is important to inform the
laboratory that both lipid-independent and lipid-dependent species are of clinical
interest. M. pachydermatis is unique in that it grows well in Sabouraud dextrose
agar at 32 °C to 37 °C without lipid supplementation; however, lipid-dependent
Malassezia species will not grow on Sabouraud dextrose agar. Modified Dixon
Agar and Leeming medium support growth of all Malassezia species. Due to the
presence of lipid-dependent yeasts on the skin of cats, the use of lipid-supplemented media, especially the modified Dixon’s medium or Leeming medium, is
required [8, 29, 30].
PCR
PCR is not used in the routine diagnosis of Malassezia dermatitis in cats. Culture-­
based methods do not always allow species-specific identification and, if this is
necessary, polymerase chain reaction (PCR) is a viable option. PCR uses laboratory
methods that amplify DNA from a sample, even directly from skin or from culture
with high accuracy and efficiency [31]. Recent findings show that the multiplex-­
real-­time PCR was highly effective in identifying Malassezia species from animal
and human samples [32].
Malassezia
355
VetBooks.ir
Histopathology and Skin Biopsy
Skin biopsy is not routinely used to diagnose Malassezia dermatitis in cats. If the
cat is otherwise healthy and yeast are noted on the skin biopsy, their presence is
most likely due to the underlying skin disease, e.g., hypersensitivity disease or
primary disorder of keratinization. However, if the cat is ill and has marked skin
lesions, the presence of Malassezia organism should be interpreted as a sign of
systemic illness and a thorough medical evaluation pursued. Histological sections
reporting Malassezia yeasts often note their presence in the stratum corneum of
the epidermis or follicular infundibulum [26]. In cases of severe exfoliation, they
may be reported in areas of mild to severe orthokeratotic to parakeratotic hyperkeratosis [26].
Treatment
Treatment of Malassezia dermatitis in cats is individualized and depends on the
severity of clinical signs and potential underlying cause. If the underlying cause is
not identified and treated, Malassezia dermatitis will not resolve. If the underlying
disease is chronic, e.g., hypersensitivity dermatitis, the owner should be warned that
disease flares will cause relapses of Malassezia dermatitis.
Topical Therapy
The major obstacle to topical therapy for Malassezia dermatitis in cats is what
the cat and owner can tolerate. Ideally, topical therapy is the treatment of choice.
Attention to coat hygiene is important if there is matting or retained hairs. If the
cat will tolerate bathing, the topical shampoos of choice are miconazole/chlorhexidine, ketoconazole/chlorhexidine, or climbazole/chlorhexidine combinations once
or twice weekly. The authors have found it very helpful for owners to understand
that Malassezia dermatitis is commonly associated with bacterial overgrowth, so
combination products are the best choice. If the cat is otherwise healthy but has generalized lesions, whole body bathing is recommended. Given that this may not be
possible, other options include the use of leave-on mousse products with the above
ingredients. If lesions are focal, these combination products can be applied to just
the affected areas. It is important to remember that grooming activities of cats put
them at greater risk of adverse reactions to topical products.
Systemic Antifungal Therapy
Oral antifungal therapy is indicated if the topical therapy is impractical or ineffective. The oral antifungal of choice is itraconazole (Itrafungol, Elanco Animal
Health). It is labelled for use in cats at 5 mg/kg orally once daily on an alternating
VetBooks.ir
356
M. L. Piccione and K. A. Moriello
week on/week off treatment schedule for dermatophytosis [33]. Itraconazole is generally well tolerated in cats and safe at this dose. The most common side effects
reported were hypersalivation, decreased appetite, vomiting and diarrhea [33]. It is
important to stress to clients that compounded itraconazole should not be used as
there is strong evidence that it is not bioavailable [34].
The efficacy of oral itraconazole for treatment of Malassezia dermatitis was
reported in two studies. In a retrospective study, 15 cats received 5 to 10 mg/kg
itraconazole (Itrafungol/Janssen), administered orally once daily as the sole therapy
[35]. Affected cats had either localized (n = 8) or generalized lesions (n = 7). Twelve
of the cats had concurrent otitis externa. Itraconazole was effective in all cats with
no reported side effects. In another study, pulse therapy (week on/week off) itraconazole was used to treat Malassezia dermatitis in 6 Devon Rex cats with concurrent
seborrheic dermatitis [36]. There was a marked improvement in clinical signs with
a reduction of inflammation and pruritus.
Yeast Otitis
Malassezia is a common cause of otitis externa in cats (Chapter, Otitis). It is most
common in cats with hypersensitivity dermatitis (Fig. 2). Immediate treatment may
include systemic itraconazole if there are large numbers of yeasts present and pruritus is severe. However, in most cases Malassezia otitis can be managed with weekly
ear cleaning and topical application of an otic antifungal/glucocorticoid product.
Long-term management of Malassezia otitis externa can be done successfully with
once or twice weekly application of otic steroids. The authors frequently compound
equal portions of injectable dexamethasone ear drops in saline or propylene glycol
for the owner to apply. This avoids the unnecessary use of antimicrobials when the
primary need is just an anti-inflammatory action.
Zoonotic Implications
Malassezia organisms are found on both people and animals. Malassezia pachydermatis is not a normal commensal organism. However, since cats can be colonized
by both lipid-independent and lipid-dependent organisms, it is important for veterinary health care workers to practice good hand hygiene when handling cats and to
remind owners to do the same.
Conclusion
Feline Malassezia dermatitis is a superficial fungal skin disease that can present
with a wide range of clinical signs. Clinical signs are caused by overgrowth of normal body flora, and concurrent overgrowth of bacteria is common. Malassezia dermatitis is commonly associated with chronic skin diseases such as hypersensitivity
VetBooks.ir
Malassezia
357
disorders, seborrhea, and underlying metabolic diseases that can trigger changes
in the skin immune system. Devon Rex cats appear particularly susceptible to both
Malassezia colonization and M. pachydermatis associated seborrheic dermatitis,
without evidence of systemic disease. Cytological examination is the most useful
technique for assessment of the density of Malassezia yeasts on the skin surface.
Additionally, contact-plate fungal cultures also provide a convenient technique for
isolation and quantification of yeast colonies. PCR allows for rapid identification
and speciation of samples analyzed as well. M. pachydermatis is the main species
identified in cats, but lipid-dependent species, particularly in the claw folds, can
also be found. Itraconazole is the systemic drug of choice along with concurrent
topical therapy. Although rare, the finding of Malassezia dermatitis in cats with
widespread skin lesions should prompt the clinician to consider whether or not this
is an early marker of systemic disease.
References
1. Theelen B, Cafarchia C, Gaitanis G, et al. Malassezia ecology, pathophysiology, and treatment. Med Mycol. 2018;56:S10–25.
2. Cabañes FJ. Malassezia yeasts: how many species infect humans and animals? PLoS Pathog.
2014;10:e1003892.
3. Colombo S, Nardoni S, Cornegliani L, et al. Prevalence of Malassezia spp. yeasts in feline nail
folds: a cytological and mycological study. Vet Dermatol. 2007;18:278–83.
4. Volk AV, Belyavin CE, Varjonen K, et al. Malassezia pachydermatis and M nana predominate
amongst the cutaneous mycobiota of Sphynx cats. J Feline Med Surg. 2010;12:917–22.
5. Bond R, Howell S, Haywood P, et al. Isolation of Malassezia sympodialis and Malassezia
globosa from healthy pet cats. Vet Rec. 1997;141:200–1.
6. Crespo M, Abarca M, Cabanes F. Otitis externa associated with Malassezia sympodialis in two
cats. J Clin Microbiol. 2000;38:1263–6.
7. Guillot J, Gueho E, Lesord M, et al. Identification of Malassezia species: a practical approach.
J Mycol Med. 1996;6:103–10.
8. Böhmová E, Čonková E, Sihelská Z, et al. Diagnostics of Malassezia Species: a review. Folia
Vet. 2018;62:19–29.
9. Tai-An C, Hill PB. The biology of Malassezia organisms and their ability to induce immune
responses and skin disease. Vet Dermatol. 2005;16:4–26.
10. Buommino E, De Filippis A, Parisi A, et al. Innate immune response in human keratinocytes
infected by a feline isolate of Malassezia pachydermatis. Vet Microbiol. 2013;163:90–6.
11. Sparber F, LeibundGut-Landmann S. Host responses to Malassezia spp. in the mammalian
skin. Front Immunol. 2017;8:1614.
12. Åhman SE, Bergström KE. Cutaneous carriage of Malassezia species in healthy and seborrhoeic Sphynx cats and a comparison to carriage in Devon Rex cats. J Feline Med Surg.
2009;11:970–6.
13. Bond R, Stevens K, Perrins N, et al. Carriage of Malassezia spp. yeasts in Cornish Rex, Devon
Rex and Domestic short-haired cats: a cross-sectional survey. Vet Dermatol. 2008;19:299–304.
14. Nardoni S, Mancianti F, Rum A, et al. Isolation of Malassezia species from healthy cats and
cats with otitis. J Feline Med Surg. 2005;7:141–5.
15. Crespo M, Abarca M, Cabanes F. Occurrence of Malassezia spp. in the external ear canals of
dogs and cats with and without otitis externa. Med Mycol. 2002;40:115–21.
16. Dizotti C, Coutinho S. Isolation of Malassezia pachydermatis and M. sympodialis from the
external ear canal of cats with and without otitis externa. Acta Vet Hung. 2007;55:471–7.
VetBooks.ir
358
M. L. Piccione and K. A. Moriello
17. Åhman S, Perrins N, Bond R. Carriage of Malassezia spp. yeasts in healthy and seborrhoeic
Devon Rex cats. Sabouraudia. 2007;45:449–55.
18. Bond R, Curtis C, Ferguson E, et al. An idiopathic facial dermatitis of Persian cats. Vet
Dermatol. 2000;11:35–41.
19. Ordeix L, Galeotti F, Scarampella F, et al. Malassezia spp. overgrowth in allergic cats. Vet
Dermatol. 2007;18:316–23.
20. Meason-Smith C, Diesel A, Patterson AP, et al. Characterization of the cutaneous mycobiota
in healthy and allergic cats using next generation sequencing. Vet Dermatol. 2017;28:71–e17.
21. Pressanti C, Drouet C, Cadiergues M-C. Comparative study of aural microflora in healthy cats,
allergic cats and cats with systemic disease. J Feline Med Surg. 2014;16:992–6.
22. Sierra P, Guillot J, Jacob H, et al. Fungal flora on cutaneous and mucosal surfaces of cats infected
with feline immunodeficiency virus or feline leukemia virus. Am J Vet Res. 2000;61:158–61.
23. Perrins N, Gaudiano F, Bond R. Carriage of Malassezia spp. yeasts in cats with diabetes mellitus, hyperthyroidism and neoplasia. Med Mycol. 2007;45:541–6.
24. Hljfftee Ma M-V, Curtis C, White R. Resolution of exfoliative dermatitis and Malassezia
pachydermatis overgrowth in a cat after surgical thymoma resection. J Small Anim Pract.
1997;38:451–4.
25. Godfrey D. A case of feline paraneoplastic alopecia with secondary Malassezia associated
dermatitis. J Small Anim Pract. 1998;39:394–6.
26. Mauldin EA, Morris DO, Goldschmidt MH. Retrospective study: the presence of Malassezia
in feline skin biopsies. A clinicopathological study. Vet Dermatol. 2002;13:7–14.
27. Toma S, Cornegliani L, Persico P, et al. Comparison of 4 fixation and staining methods for the
cytologic evaluation of ear canals with clinical evidence of ceruminous otitis externa. Vet Clin
Pathol. 2006;35:194–8.
28. Griffin JS, Scott D, Erb H. Malassezia otitis externa in the dog: the effect of heat-fixing otic
exudate for cytological analysis. J Veterinary Med Ser A. 2007;54:424–7.
29. Guillot J, Bond R. Malassezia pachydermatis: a review. Med Mycol. 1999;37:295–306.
30. Peano A, Pasquetti M, Tizzani P, et al. Methodological issues in antifungal susceptibility testing of Malassezia pachydermatis. J Fungi. 2017;3:37.
31. Vuran E, Karaarslan A, Karasartova D, et al. Identification of Malassezia species from pityriasis versicolor lesions with a new multiplex PCR method. Mycopathologia. 2014;177:41–9.
32. Ilahi A, Hadrich I, Neji S, et al. Real-time PCR identification of six Malassezia species. Curr
Microbiol. 2017;74:671–7.
33. Puls C, Johnson A, Young K, et al. Efficacy of itraconazole oral solution using an alternating-­
week pulse therapy regimen for treatment of cats with experimental Microsporum canis infection. J Feline Med Surg. 2018;20:869–74.
34. Mawby DI, Whittemore JC, Fowler LE, et al. Comparison of absorption characteristics of oral
reference and compounded itraconazole formulations in healthy cats. J Am Vet Med Assoc.
2018;252:195–200.
35. Bensignor E. Treatment of Malassezia overgrowth with itraconazole in 15 cats. Vet Rec.
2010;167:1011–2.
36. Åhman S, Perrins N, Bond R. Treatment of Malassezia pachydermatis-associated seborrhoeic
dermatitis in Devon Rex cats with itraconazole–a pilot study. Vet Dermatol. 2007;18:171–4.
VetBooks.ir
Viral Diseases
John S. Munday and Sylvie Wilhelm
Abstract
Viruses are becoming increasingly recognized as an important cause of feline
skin disease. Diseases associated with viruses in cats include hyperplastic and
neoplastic skin disease caused by papillomaviruses, erosive and ulcerative skin
disease caused by herpesviruses and poxviruses, and skin lesions that develop as
a part of a more generalized viral infection as is seen due to calicivirus infection.
Skin disease may also be seen in cats infected by feline leukemia virus and feline
infectious peritonitis virus. In this chapter, the etiology and epidemiology of
infection by each of the viruses that cause feline skin disease are reviewed along
with the clinical disease presentation, the histological lesions, and other appropriate diagnostic tests. Additionally, the expected clinical course of the diseases
and the currently recommended therapies are described.
Introduction
Viruses have traditionally been thought to rarely cause skin disease in cats. However,
research in the last 30 years has expanded both the number of viruses that cause
feline skin disease and the types of skin lesions caused by these viral infections.
Feline viral infections can be broadly subdivided into those that cause hyperplastic or neoplastic skin disease (papillomaviruses), those that cause cell lysis and
generally self-resolving inflammatory disease (herpesvirus, poxvirus), and those
that infrequently cause skin lesions as part of a more generalized viral infection
J. S. Munday (*)
Massey University, Palmerston North, New Zealand
e-mail: j.munday@massey.ac.nz
S. Wilhelm
Vet Dermatology GmbH, Richterswil, Switzerland
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_17
359
VetBooks.ir
360
J. S. Munday and S. Wilhelm
(calicivirus, feline leukemia virus, feline infectious peritonitis virus). While feline
immunodeficiency virus is briefly discussed, it is currently uncertain whether or not
this virus causes skin disease in cats.
Papillomaviruses
Papillomaviruses (PVs) are small, non-enveloped, circular double-stranded DNA
viruses that typically infect stratified squamous epithelium. Their DNA contains
seven open reading frames (ORFs), including five that code for the early (E) proteins and two that code for the late (L) proteins [1]. Their life cycle is dependent on
the terminal differentiation, keratinization, and desquamation of epithelial cells, and
feline PVs cause disease due to the ability of their E7 proteins to alter the normal
growth and differentiation of these cells. Papillomaviruses are considered one of the
oldest viral families and have co-evolved with their hosts over a long time. For this
reason, the majority of PVs are species-specific and the overwhelming majority of
PV infections are asymptomatic [2].
Papillomaviruses are classified by comparing the similarities of the L1 ORF [3].
Currently five PV types are recognized to infect cats, including Felis catus papillomavirus (FcaPV) type 1, which is classified in the Lambdapapillomavirus genus
[4, 5]; FcaPV-2, which is classified in the Dyothetapapillomavirus genus [6]; and
FcaPV-3, -4 and -5, which have not been fully classified, but will likely be grouped
together in a novel feline PV genus [7–9].
Although most PV infections are asymptomatic, PVs were first proposed to be a
cause of feline skin disease in 1990, when PV-induced cell changes were observed
in a cutaneous plaque [10]. Since this time, the importance of PVs as a cause of
skin disease has been increasingly recognized and PVs are currently thought to
cause viral plaques/Bowenoid in situ carcinomas, a proportion of squamous cell
carcinomas, feline sarcoids, a proportion of basal cell carcinomas and cutaneous
viral papillomas [11].
Feline Viral Plaques/Bowenoid In Situ Carcinoma
These lesions have traditionally been thought of as two separate skin diseases of
cats. However, as viral plaques and Bowenoid in situ carcinomas (BISCs) share
many histological features and transitional lesions between the two lesions are often
visible [12], it appears these two lesions are different severities of the same process.
Etiology and Epidemiology
FcaPV-2 is thought to be the predominant cause of these lesions [13, 14]. Current
evidence suggests most kittens are infected from the dam within the first few weeks
of life [15]. Infection by FcaPV-2 is probably lifelong and often does not stimulate
an antibody response [16]. As most cats are infected by FcaPV-2, but few develop
viral plaques/BISCs, it appears that host factors are important in determining
VetBooks.ir
Viral Diseases
361
whether or not a cat will develop clinical disease. While immunosuppressed cats
may be at increased risk of viral plaque/BISC development, many cats have been
reported to develop lesions without any detectable immunosuppression, and the factors that predispose a cat to lesion development are largely unknown [17]. The early
development and severe manifestation of viral plaques/BISCs in Devon Rex and
Sphinx cats suggests a genetic susceptibility, although the basis of this susceptibility is unknown [18]. Viral plaques/BISCs have also been associated with infection
by FcaPV-3 and FcaPV-5. Currently little is known about the epidemiology of these
viruses.
Clinical Presentation
Viral plaques/BISCs most often develop between the ages of eight and 14 years,
although they have been reported in cats as young as 7 months of age [12, 19]. Cats
with viral plaques tend to be younger than those with BISCs, supporting the hypothesis that some viral plaques progress to BISCs. Viral plaques most often develop on
the trunk, head, or neck although in advanced cases lesions can develop anywhere
on the body. They are often multiple and small, generally less than 1 cm in diameter,
scaly papules or plaques that may be either pigmented or nonpigmented and can be
covered by thin crusts (Fig. 1). While BISCs can appear clinically very similar to
viral plaques, they tend to be larger, more markedly raised, and can be ulcerated or
covered by a serocellular crust or a thick layer of keratin (Fig. 2). The head, neck,
and limbs are most commonly affected. Viral plaques and BISCs can develop within
pigmented or nonpigmented, haired or nonhaired skin and neither lesion is typically
painful or pruritic [12].
Histopathology and Diagnosis
Histology of a viral plaque reveals a well-demarcated focus of mild epidermal hyperplasia. Cells retain their orderly maturation and no dysplasia is visible
(Fig. 3). Histology of a BISC reveals a well-demarcated focus of marked epidermal
Fig. 1 Feline viral plaque.
Plaques most frequently
appear as focal, slightly
raised lesions around the face
of cats. Feline viral plaques
and Bowenoid in situ
carcinomas appear to be
different severities of the
same disease process with
viral plaques the milder form
of the disease. (Courtesy of
Dr. Sharon Marshall,
Veterinary Associates,
Hastings, New Zealand)
J. S. Munday and S. Wilhelm
VetBooks.ir
362
Fig. 2 Feline Bowenoid in situ carcinoma. As with viral plaques, these often develop on the head
of cats. Compared to a viral plaque, Bowenoid in situ carcinomas are larger, more markedly raised,
and covered by increased quantities of keratin. However, as viral plaques and Bowenoid in situ
carcinomas represent different severities of the same disease process and there is no clear distinction between the two lesions. (Courtesy of Dr. Richard Malik, Centre for Veterinary Education,
University of Sydney, Australia)
Fig. 3 Feline viral plaque.
Plaques appear as
well-demarcated foci of
mild to moderate
epidermal hyperplasia.
Little dysplasia is visible
within the hyperplastic
cells and orderly
maturation of the cells is
retained (HE, 200×)
hyperplasia that can extend to involve follicular infundibula. The hyperplastic cells
can form well-demarcated solid masses of basilar cells that bulge into the underlying dermis. Examination of deeper layers of the BISC reveals keratinocyte dysplasia with crowding of basal cells and cells with nuclei that are elongated vertically
(windblown cells) [20]. Dyskeratosis is rarely visible within BISCs. Although significant atypia can be present, the cells remain confined by the basement membrane
(Fig. 4). Viral replication can result in prominent PV-induced changes. However,
keratinocyte dysplasia can prevent viral replication and PV-induced cell changes
363
VetBooks.ir
Viral Diseases
Fig. 4 Feline Bowenoid in situ carcinoma. Compared to the viral plaque, the hyperplasia is more
marked with prominent involvement of follicular infundibula. There is moderate atypia within the
cell population, but no penetration of the basement membrane. While papillomavirus-induced cell
changes are prominent in this lesion, more advanced Bowenoid in situ carcinomas often contain
little histological evidence of papillomavirus infection. (HE, 200×)
Fig. 5 Feline viral plaque.
Papillomavirus-induced
cell changes include the
presence of keratinocytes
that have dark nuclei
surrounded by a clear halo
(koilocytes; arrows) as
well as the presence of
cells that contain increased
quantities of grey-blue
smudged cytoplasm
(arrowheads; HE, 400×)
are rare in larger more developed BISCs. PV-induced changes include the presence
of large keratinocytes with clear or blue-grey granular cytoplasm and/or shrunken
nuclei that are surrounded by a clear halo (koilocytes; Fig. 5) [17]. Eosinophilic
intranuclear inclusions can be visible, although care has to be taken to differentiate
these from nucleoli. Hyperplasia of cells deeper within follicles or hyperplasia of
sebaceous glands may be visible in viral plaques/BISCs that are caused by FcaPV-3
or -5. Additionally, these lesions contain prominent basophilic cytoplasmic inclusions that are often flattened against the nucleus [8, 21]. If no PV-induced changes
are visible, then differentiation from actinic in situ carcinoma (actinic keratosis) is
VetBooks.ir
364
J. S. Munday and S. Wilhelm
required. Features that support a BISC rather than an actinic lesion include the consistently altered nuclear polarity of the basal cells, the sharp demarcation between
affected and normal epidermis, and the follicular involvement. In addition, actinic
lesions will also often have solar elastosis visible in the underlying dermis.
Immunohistochemistry can be used in cases in which histological differentiation
between a Bowenoid and an actinic in situ carcinoma is problematic. Antibodies to
detect PV antigen can be used. However, antigens are only produced during viral
replication and it is rare to have immunohistochemical evidence of PV infection in a
lesion that does not contain PV-induced cell changes [17]. Therefore, p16CDK2NA protein (p16) immunohistochemistry is recommended to investigate a PV etiology. The
detection of a marked increase in p16 suggests a PV etiology because PVs cause cell
dysregulation by mechanisms that consistently increase p16 (Fig. 6). In contrast, in
actinic lesions, loss of cell regulation is caused by mechanisms that do not increase
p16 [22]. When performing p16 immunohistochemistry it has to be remembered
that only the G175-405 human p16 clone has been shown to cross-­react with the
feline p16 protein. As p53 immunostaining can be present in both actinic keratosis and BISCs, this will not be useful to differentiate between a Bowenoid and an
actinic lesion [22]. Due to the frequency with which PVs asymptomatically infect
skin, the detection of PV DNA in a lesion does not confirm a diagnosis of BISC or
exclude a diagnosis of actinic in situ carcinoma.
Treatment
Viral plaques and BISCs can spontaneously resolve, persist without progressing, or
slowly increase in size and number. In addition, all viral plaques/BISCs should be
carefully monitored for progression to a SCC. Lesions in Devon Rex and Sphinx
cats can rapidly progress to SCCs that have metastatic potential [18, 23].
Surgical excision of a viral plaque or BISC is expected to be curative, although
additional lesions may subsequently develop at different locations. Imiquimod
cream has been used to treat genital warts in people and has been suggested as a
Fig. 6 Feline viral plaque.
The use of antibodies
against p16CDKN2A protein
reveals intense nuclear and
cytoplasmic
immunostaining
throughout the hyperplastic
epidermis (Hematoxylin
counterstain 400×)
VetBooks.ir
Viral Diseases
365
possible treatment. Imiquimod stimulates toll-like receptors and locally increases
alpha interferon and tumor necrosis factor-α [24]. It is a topical therapy, usually
applied three times per week for 8–16 weeks. In an uncontrolled study of 12 cats
with BISCs, imiquimod resulted in partial resolution of at least one BISC in all
12 cats and complete remission of at least one BISC in 5 cats [25]. Side effects
included local erythema and mild discomfort in five cats and potential signs of
systemic toxicity, including neutropenia, elevated hepatic enzymes, anorexia, and
weight loss were observed in two cats. While there is anecdotal evidence supporting
the use of imiquimod cream, additional controlled studies are required to determine
the efficacy and safety of this treatment. In humans, imiquimod has also been used
to treat basal cell carcinomas and actinic lesions and this treatment does not appear
to have a specific action against PV-induced lesions. Imiquimod is currently not
recommended as a primary treatment for pre-neoplastic or neoplastic skin lesions
in people, but may be effective if better established therapies are not available [26].
Likewise, in veterinary medicine, imiquimod has been used to treat viral and nonviral in situ carcinomas when other treatments were considered impractical, and
investigation of a PV etiology may not be necessary prior to the use of imiquimod.
Photodynamic therapy could be another treatment option as excellent response rates
were recently reported, although there was no attempt in this study to differentiate
between PV-induced and actinic in situ carcinomas [27].
Autologous vaccination has not been evaluated as a method of treating viral
plaques/BISCs in cats. However, considering the immune response to a PV-induced
lesion, this treatment modality is not expected to work. There is currently little
evidence from any species that vaccination using autologous or virus-like particle
vaccines has any significant efficacy in treating either PV-induced warts or preneoplastic lesions.
Cutaneous Squamous Cell Carcinomas
Squamous cell carcinomas (SCCs) are one of the most common skin neoplasms
of cats and are a significant cause of morbidity and mortality (Chapter, Genetic
Diseases for more information). While there can be no doubt that solar exposure is
a significant cause of SCCs, there is evidence that PVs may also contribute to the
development of some neoplasms. Evidence of a role of PVs includes the detection
of FcaPV-2 DNA more frequently in cutaneous SCCs than in non-SCC skin samples
[13]. Additionally, p16 immunostaining is visible within SCCs that contain PV DNA
(Fig. 7) and SCCs that have p16 immunostaining demonstrate a different biological
behavior, suggesting that they may have been caused by different carcinogenic pathways [28, 29]. Furthermore, FcaPV-2 RNA can be detected in a proportion of feline
cutaneous SCCs and the proteins that are expressed by FcaPV-2 have been shown
to have transforming properties in cell cultures [30, 31]. Overall current evidence
suggests that PV infection causes most SCCs that develop in haired, pigmented skin
and that PV infection, probably with UV light as a co-­factor, could promote the
development of between a third and a half of SCCs from nonhaired, nonpigmented
VetBooks.ir
366
J. S. Munday and S. Wilhelm
Fig. 7 Cutaneous
squamous cell carcinoma.
Immunostaining for
p16CDKN2A protein is
diffusely present within the
nucleus and cytoplasm of
the neoplastic cells.
Papillomavirus DNA was
amplified from this
neoplasm using PCR
(Hematoxylin counterstain
200×)
skin [29]. However, as asymptomatic infection of the skin is extremely common in
cats, it is currently impossible to definitively determine that role that FcaPV-2 plays
in the development of cutaneous SCCs in cats.
Feline Sarcoids
Feline sarcoids are rare neoplasms in cats. They have also been called “fibropapillomas”; however, as fibropapillomas are considered hyperplastic rather than neoplastic lesions, the term “sarcoid” is preferred.
Etiology and Epidemiology
Bovine papillomavirus (BPV) type 14 has been consistently detected in feline
sarcoids throughout the world [32–34], and infection by BPV-14 is thus considered to be the cause of this disease. BPV-14 is a Deltapapillomavirus that is
most closely related to BPV-1 and -2, the causes of equine sarcoids [35]. The
bovine deltapapillomaviruses have the unique ability to cause both self-resolving fibropapillomas in cattle and mesenchymal neoplasia in non-host species.
Cows are commonly asymptomatically infected by BPV-14 [36], but BPV-14
was not detected in a large number of cutaneous and oral samples from cats
[32]. This suggests that cats are probably dead-end hosts for the PV. It is currently unknown how BPV-14 is transmitted from cattle to cats. However, as this
disease appears to be most common in cats that live in dairy barns, close contact
with cattle appears to be necessary. It is also unknown whether any co-factors
are required to allow BPV-14 to cause sarcoids. Evidence from horses suggests
that mesenchymal cell proliferation may be important for equine sarcoid development and it is possible that cat fight wounds could be important in allowing
introduction of the PV into the dermis and stimulating dermal mesenchymal
proliferation.
VetBooks.ir
Viral Diseases
367
Fig. 8 Feline sarcoid. The
mass protrudesi from close
to the nasal philtrum of
this cat. (Courtesy of Dr.
William Miller, Cornell
University College of
Veterinary Medicine,
Ithaca, New York)
Clinical Presentation
Feline sarcoids have only been reported in outdoor cats from rural environments and
are most common in younger male cats. They develop as solitary or multiple slow-­
growing exophytic nonulcerated nodules most frequently around the face, especially involving the nasal philtrum and upper lip, although sarcoids have also been
reported in distal limbs and tail (Fig. 8) [33]. There is some evidence to suggest
feline sarcoids may also rarely develop within the oral cavity.
Histopathology and Diagnosis
A feline sarcoid should be suspected if an exophytic mass is observed around the
mouth or nose of a young cat that has contact with cattle. Unlike typical PV infections of the skin, infection by the PV is confined to the dermis [34]. Therefore,
the predominant histological feature of a sarcoid is a proliferation of moderately
well-­differentiated mesenchymal cells within the dermis (Fig. 9). The proliferative
dermal mass is covered by hyperplastic epidermis that extends into the mesenchymal cells by the formation of prominent rete pegs [33, 34]. As the sarcoid does not
support viral replication, sarcoids do not contain any PV-induced cell changes and
immunohistochemistry will not reveal the presence of PV L1 antigen [34]. The
amplification of BPV-14 DNA from the lesion confirms a diagnosis of feline sarcoid.
Treatment
While there are few clinical reports of feline sarcoids, these neoplasms appear to
be locally infiltrative, but do not metastasize. In the authors’ experience, complete
surgical excision is curative. However, as these lesions often develop around the
nose and mouth, complete excision can be problematic and feline sarcoids often
recur and show an increased growth rate after surgery. A cat with recurrent sarcoids
was treated with topical imiquimod and intralesional cisplatin, but neither treatment
appeared to alter the disease course and the cat was eventually euthanatized due to
the local effects of the neoplasm [35].
VetBooks.ir
368
J. S. Munday and S. Wilhelm
Fig. 9 Feline sarcoid. The
neoplasm consists of
moderately welldifferentiated fibroblasts
that are covered by
hyperplastic epidermis that
forms prominent rete pegs
(HE, 200×)
Basal Cell Carcinomas
These are rare neoplasms and only a limited number have been assessed for a
PV etiology. However, a potential role of PVs in the development of feline basal
cell carcinomas (BCCs) is supported by the observation that a proportion contain
PV-induced changes [20, 37, 38]. Feline BCCs have not been reported to be caused
by FcaPV-2. Instead feline BCCs have been associated with FcaPV-3 and a novel
unclassified PV type [37, 38].
Cutaneous Papillomas
In cats, FcaPV-1 causes oral papillomas that typically develop on the ventral surface
of the tongue [39]. There are also sporadic reports of cutaneous viral papillomas.
While cross-species infection by a human PV was originally suspected [40], this
appears unlikely and spread of FcaPV-1 from the mouth to the skin of cats appears
to be a more likely cause of these rare lesions.
Herpesviruses
Feline herpesvirus 1 is a double-stranded DNA Alphaherpesvirus that is a common
cause of upper respiratory tract disease and conjunctivitis in younger cats. In 1971,
it was reported that herpesvirus infection may also cause dermatitis in cats [41], and
feline herpesvirus dermatitis is now recognized as a distinct, albeit rare, manifestation of herpesvirus infection.
Etiology and Epidemiology
The rate of feline herpesvirus 1 infection is difficult to determine as many cats
are vaccinated against this virus at an early age. Infection of an unvaccinated cat
VetBooks.ir
Viral Diseases
369
typically results in clinical signs of upper respiratory disease, such as rhinotracheitis, and conjunctivitis. While the clinical signs usually resolve within a few days
or weeks, the herpesvirus infection can become latent, especially in the trigeminal
ganglia. These latent infections can become recrudescent if the cat becomes immunosuppressed. It is hypothesized that recrudescence of previous herpesviral infections within the cutaneous nerves could cause feline herpesvirus dermatitis [42].
Due to the likely role of immunosuppression in the pathogenesis of disease, cats
receiving glucocorticoids may be predisposed to disease development [42]. Cats
that are in a household with numerous other cats also appear to be at increased risk,
although it is uncertain whether this is because the cats are immunosuppressed due
to stress or because the cats are more likely to be exposed to herpesvirus [42]. Feline
herpesvirus dermatitis has not been associated with infection by feline immunodeficiency virus or feline leukemia virus. While previous infection by herpesvirus
is thought to be key in the pathogenesis of this disease, herpesvirus dermatitis has
been reported in cats that have a good vaccination history and in cats that do not
have a history of previous respiratory disease [43].
Clinical Presentation
Herpesvirus dermatitis appears to be most common in cats around 5 years of age
although this disease has been reported in cats 4 months to 17 years old [42, 44].
Most cats with herpesvirus dermatitis have lesions almost exclusively on the face
with the dorsal muzzle to the bridge of the nose and periocular skin most frequently
affected (Fig. 10). The lips can also be affected and, in rare cases, the lesions can
become generalized over the body within a few days [42, 43, 45]. Lesions are typically erosions and ulcers that are covered by a thick serocellular crust and have
been referred to as “ulcerative facial and nasal dermatitis and stomatitis syndrome.”
The lesions tend to be roughly spherical and are often asymmetrical, but the development of symmetrical lesions does not exclude a herpesviral etiology. Regional
lymphadenopathy can be present [45]. Oral lesions are only rarely reported in cats
Fig. 10 Feline herpesviral
dermatitis. This disease
typically presents as
multiple ulcerative lesions
over the face, especially
around the bridge of the
nose. (Courtesy of Dr.
Richard Malik, Centre for
Veterinary Education,
University of Sydney,
Australia)
VetBooks.ir
370
J. S. Munday and S. Wilhelm
with herpesvirus dermatitis [45], and affected cats may or may not have active or
historical evidence of respiratory disease. The skin lesions can be intensely pruritic,
thus mimicking a wide range of possible differential diagnoses, especially in the
absence of respiratory signs of disease. Depending on the localization of the lesions,
differential diagnoses include allergic skin disease, calicivirus-associated dermatitis, autoimmune skin diseases, and erythema multiforme.
Exfoliative erythema multiforme is a rare disease that has been reported to
develop following infection by herpesvirus. Clinical signs include widespread scaling (exfoliation) in combination with alopecia. Accompanying systemic symptoms
are possible and the lesions spontaneously resolve after clearance of the herpesvirus
infection [46].
Histopathology and Diagnosis
Histology of a lesion reveals full-thickness necrosis and loss of the epidermis.
Underlying the areas of necrosis there are typically large numbers of inflammatory cells including a high proportion of eosinophils (Fig. 11). Necrosis of the epithelium can extend into the underlying follicular infundibula and adnexal glands.
The epidermis adjacent to the areas of ulceration can be thickened and spongiotic.
The lesions are covered by a marked serocellular crust that consists of degenerate
inflammatory cells and fibrin. Careful examination of the intact epidermis adjacent
to the necrosis, the follicles, and the adnexal glands may reveal the rare presence
of intranuclear viral inclusions (Fig. 12). These are eosinophilic and surrounded by
marginated nuclear material. Making a definitive diagnosis should not be problematic when inclusion bodies are present. However, in cases that do not have visible
inclusions, additional techniques may be necessary. The most conclusive evidence
supporting a diagnosis is the demonstration of herpesviral antigens within the
lesions using immunohistochemistry [44]. The failure to amplify herpesviral DNA
Fig. 11 Feline herpesviral
dermatitis. The dermis
contains large numbers of
inflammatory cells
including a large
proportion of eosinophils
(HE, 200×)
VetBooks.ir
Viral Diseases
371
Fig. 12 Feline herpesviral
dermatitis. The epidermis
adjacent to areas of
ulceration is thickened and
spongiotic with some cells
containing eosinophilic
intranuclear viral
inclusions (arrows; HE,
400×)
from a lesion using PCR excludes a diagnosis of herpesviral dermatitis. However, as
DNA from latent or vaccinal herpesvirus infection or contamination from infected
mucosa from grooming can be detected by PCR, this technique cannot be used to
confirm a diagnosis of herpesviral dermatitis [47, 48]. Persico et al recently recommended that PCR can be used as a screening test for cases in which no viral inclusions are present, but immunohistochemistry was required to confirm a diagnosis of
herpesviral dermatitis [48].
Treatment
Herpesvirus dermatitis may resolve spontaneously, although as few untreated cats
are described in the literature, the frequency of self-cure is uncertain. In some cats,
supportive care such as treatment of secondary bacterial infection may result in
resolution of the clinical signs of disease [43]. As immunosuppression could contribute to disease development, any immunosuppressive treatments should be discontinued. Small lesions can be surgically excised, although whether or not the
lesions would have spontaneously resolved if they had been left is unknown [42].
While herpesviral skin disease in humans typically spontaneously resolves, numerous treatments have been developed to accelerate disease resolution. Some of these
antiviral drugs may also be beneficial in cats, but they generally have complicated
pharmacokinetics that may render them ineffective or toxic in cats and none has
consistently been found to be safe and effective [49, 50]. Currently, famciclovir
has the greatest amount of evidence supporting efficacy, both in naturally infected
cats and in a placebo-controlled study of experimentally infected cats. The used
dosages vary from 40 to 90 mg/kg once or twice daily to 125 mg/kg every 8 hrs
[51, 52]. Topical “cold sore” creams, especially those containing pencivovir may
also be beneficial and can be used with systemic famciclovir therapy (R. Malik,
pers. comm). Interferons (IFNs), including IFNα and recombinant IFNω [53], have
VetBooks.ir
372
J. S. Munday and S. Wilhelm
also been suggested as potential treatments, although no controlled trials have been
undertaken to assess their efficacy. Doses for IFNα again vary widely from 1MU/m2
subcutaneously three times a week to 0.01–1 MU/kg once daily for up to 3 weeks.
Most often 30 units/day have been used [54]. The effectiveness of lysine supplementation is highly controversial and no clinical benefit has been proven [50, 55].
Poxvirus
Poxviruses are large enveloped brick-shaped or oval linear double-stranded DNA
viruses. Their DNA is 130–360 kb in length and encodes 130–320 proteins [56]. Most
poxviruses are able to infect multiple species, and infection typically causes skin
lesions due to the tropism of the viruses for epidermal keratinocytes. Skin disease due
to poxviruses is rare in cats. Self-regressing proliferative skin disease due to infection
with orf virus and pseudocowpox virus (both Parapoxviruses) has been sporadically
reported in cats [57, 58]. However, the overwhelming majority of poxviral feline skin
disease is caused by cowpox virus, an Orthopoxvirus, and the remainder of this section describes disease due to cowpox virus infection. In addition to the role of cowpox
virus in diseases of cats, it also has to be noted that cats are important as a source of
cowpox infection of humans.
Etiology and Epidemiology
Cowpox virus is a poor name for this virus as the reservoir hosts appear to be small
mammals such as bank voles, short-tailed field voles, ground squirrels, and gerbils, with
cattle, humans, and cats all only rarely infected [59]. The limited geographic distribution of the reservoir hosts explains why this disease is restricted to western Eurasia with
the majority of clinical cases reported in the United Kingdom and Germany [59–61].
As cats are infected from a reservoir host during hunting, cowpox is limited to cats
that have access to a rural environment, and increased numbers of cases are seen in
autumn, due to increased hunting and greater numbers of reservoir host prey at this time
[60, 62]. Skin disease is thought to develop after a reservoir host bites the cat. Systemic
disease has also been reported, although it is uncertain if this results from inhalation of
the virus or from systemic spread of the virus from an initial skin infection.
While disease due to cowpox virus is rare in cats, exposure to the virus appears to
be much more common, with antibodies against orthopoxviruses detected in 2–17%
of cats from Western Europe [60, 63, 64]. Unsurprisingly, rates of exposure were
highest in populations of cats that had outside access and from areas in which clinical cases had been reported [60]. As behavioral factors that predispose to cowpox
exposure also predispose to infection with feline immunodeficiency virus (FIV), it
is possibly unsurprising that cats with antibodies against orthopoxvirus were more
likely to also have antibodies against FIV [64].
Viral Diseases
373
VetBooks.ir
Clinical Presentation
Lesions develop in younger to middle-aged cats that are able to come into contact
with a reservoir host species [65]. As the lesions are initiated by a rodent bite, they
typically start around the head or on the forelimbs. Lesions can subsequently spread
to ears and paws, possibly by grooming and some cats can develop widespread
lesions [62, 66].
The initial lesion is typically a single small raised ulcer covered by a serocellular crust at the site of inoculation by the rodent bite [62]. One to 3 weeks later,
additional similar lesions may develop. These start as macules and small nodules that enlarge up to 1 cm in diameter (Fig. 13). They then become ulcerated,
forming the typical crater-like skin lesion. These scab over and then gradually
dry and exfoliate within 4–5 weeks. Pruritus is variable [67]. Up to 20% of cats
may develop oral lesions, presumably due to the cat licking the skin lesions [66].
Cats that present with larger areas of dermal necrosis with extensive erythema,
edema, abscessation, and cellulitis have also been reported (Fig. 14) [67, 68].
Whether this more severe presentation represents infection by a more virulent
strain is uncertain. Cats may be transiently pyrexic and depressed during the
viremic phase 1–3 weeks after infection, but most do not demonstrate signs of
systemic disease on presentation [54, 66]. Rarely, cats may progress to develop
signs of respiratory disease that can progress to a fatal pneumonia, although
skin lesions are only variably present in cats that develop the respiratory form
of cowpox disease.
Pruritic lesions require differentiation from allergic skin diseases. Other differentials for nodular skin diseases in cats include infection by fungi or higher-order
bacteria as well as neoplastic skin disease.
Fig. 13 Feline cowpox
dermatitis. This cat
presented with numerous
raised nodules on the
forelimbs
VetBooks.ir
374
J. S. Munday and S. Wilhelm
Fig. 14 Feline cowpox
dermatitis. The lesions in
this cat progressed to
erythema, edema, and
cellulitis involving the paw
Fig. 15 Feline cowpox
dermatitis. Examination of
the dermis reveals necrosis
accompanied by large
numbers of neutrophils.
Epidermal cells are visible
scattered within the
inflammation. These cells
show evidence of
ballooning degeneration
and some have prominent
eosinophilic
intracytoplasmic viral
inclusions (arrows; HE,
200×)
Histopathology and Diagnosis
Due to the nonspecific nature of the skin lesions observed, biopsy and histology are
required for diagnosis. Examination of a lesion reveals necrosis of the epidermis
with ulceration. Examination of adjacent epidermis and within the epithelium of the
follicles will usually reveal marked ballooning degeneration (Fig. 15). The ballooning change within these cells often demonstrates the presence of prominent intracytoplasmic poxvirus inclusions. These inclusions are eosinophilic and round to
oval. The lesions are covered by a serocellular crust and the underlying dermis often
contains significant neutrophilic inflammation. Serology or electron microscopy
VetBooks.ir
Viral Diseases
375
can be used to confirm orthopoxvirus infection, but not the orthopoxvirus type.
Immunohistochemistry can be performed using monoclonal antibodies specific for
cowpox virus [69]. Alternatively, virus isolation from a fresh biopsy or scab material or amplification of viral DNA using PCR will enable a precise diagnosis to be
made [70].
Treatment
Feline cowpox dermatitis has a good prognosis with most cases resolving spontaneously [66]. In cases with extensive or numerous lesions, supportive care may be
required including the treatment of secondary bacterial infections. However, even
cats with severe skin lesions most often make a full recovery, although scarring can
occur [67]. The detection of serum antibodies against FIV does not influence the
prognosis [66].
Why some cats develop respiratory disease is unknown. Treatment with immunosuppressive doses of corticosteroids is contraindicated as this may predispose
to respiratory disease. While fatal pneumonia has been reported to develop in cats
that were also infected by feline leukemia virus, feline immunodeficiency virus, or
feline panleukopenia virus [66, 69], the role of these concurrent infections is uncertain and no underlying immunosuppression can be identified in most cases [61].
Cats with respiratory disease have a guarded prognosis and no specific treatments
have been shown to be beneficial. Treatment with broad spectrum antibiotics to prevent secondary bacterial infection appears to be appropriate. Additionally, four cats
with respiratory cowpox were treated with recombinant feline interferon omega.
While two of the cats survived, it is currently impossible to determine whether interferon therapy is effective in the treatment of cowpox in cats [61].
The zoonotic potential of cowpox is an important consideration when formulating a treatment plan and infected cats are thought to cause around 50% of human
cowpox infections [71]. Cowpox infection of people generally results in a transient,
focal ulcerated lesion. However, life-threatening systemic cowpox virus infections
can occur, particularly in immunosuppressed individuals [72], and in-clinic treatment may be most appropriate for cats owned by immunosuppressed clients.
Calicivirus
Feline calicivirus is a non-enveloped icosahedral single-stranded RNA virus that is
classified in the Vesivirus genus. Feline calicivirus is well recognized to cause upper
respiratory disease and oral ulcers in cats. Rarely, infection with caliciviruses can
also cause skin lesions.
376
J. S. Munday and S. Wilhelm
VetBooks.ir
Etiology and Epidemiology
Although only one calicivirus infects cats, these viruses are generally quick to
mutate and different viral strains can express different antigens and show marked
differences in virulence [73]. Feline calicivirus commonly infects cats, it is
transferred via direct contact from infected cats and is shed in ocular, nasal, and
oral secretions. Calicivirus is one of the most common viral pathogens of cats
worldwide [73]. Skin lesions can develop in association with the more typical
upper respiratory calicivirus infections. However, generalized severe skin lesions
are generally restricted to cats that develop virulent systemic disease. This rare
presentation of caliciviral disease usually develops as an outbreak, presumably
as multiple cats are exposed to a recently developed virulent calicivirus strain.
Outbreaks of virulent systemic disease have been reported involving veterinary
hospitals and animal shelters in North America and Europe, although considering the widespread distribution of this virus, outbreaks in other countries appear
likely to occur [74]. Calicivirus vaccines may reduce the severity of clinical signs
of disease, but do not appear to be protective against infection by highly virulent
calicivirus strains [75, 76].
Clinical Presentation
Acute, nonvirulent, feline calicivirus infection is usually characterized by transient,
self-limiting vesiculo-ulcerative lesions in the oral cavity (typically affecting the
tongue), on the lips and nasal philtrum. Rarely ulceration can be detected on other
body regions. Systemic signs, such as fever, depression, sneezing, conjunctivitis,
oculonasal discharge, and arthropathy resulting in lameness that resolves in a few
days (transient febrile limping syndrome) can also be observed [73].
Virulent systemic disease has been reported in cats from 8 weeks to 16 years
of age, although adult cats may be more susceptible [74, 77, 78]. Cats that present with virulent systemic disease are unwell with fever, anorexia, lethargy,
weakness, jaundice, or bloody diarrhea. Many cats will have oral ulcers [77].
Skin lesions include edema of the limbs and face with alopecia, ulcers and crusting lesions on the head (especially the lips, muzzle, and ears), and paw pads [73,
77]. Skin lesions have been reported less frequently on the abdomen and around
the anus (Fig. 16) [76].
Two cats were reported to develop pustular skin disease due to calicivirus shortly
after ovariohysterectomy. In these cats, lesions were restricted to the skin surrounding the wound. As both cats were also anorexic and depressed, it appears likely they
both had systemic virulent disease. The presence of systemic disease was supported
by the observation that one cat subsequently developed pleural effusion that necessitated euthanasia [79].
VetBooks.ir
Viral Diseases
377
Fig. 16 Virulent systemic
calicivirus infection. The
cat has ulcers and crusting
lesions on the ventral
surface of the tail and
surrounding the anus
Fig. 17 Virulent systemic
calicivirus infection.
Histology reveals
epidermal necrosis with
ulceration. The ulcers are
covered by a prominent
serocellular crust (HE,
400×)
Histopathology and Diagnosis
Histology of the skin lesions reveals ballooning degeneration and necrosis of both
surface and follicular epithelium with subsequent ulceration. Neutrophilic inflammation is often marked and ulcers are typically covered by a serocellular crust
(Fig. 17) [77]. In cases in which the epidermis remains intact, intraepidermal and
suprabasilar pustules may be visible [76]. Superficial dermal edema and vasculitis
may also be detected.
Diagnosis of systemic virulent calicivirus infection is unlikely to be made solely on
histological examination of skin samples. Instead, the skin histology will be interpreted
VetBooks.ir
378
J. S. Munday and S. Wilhelm
along with the clinical evidence of severe systemic disease to make this diagnosis.
A diagnosis of caliciviral dermatitis is supported by the demonstration of caliciviral
antigens within the lesions using immunohistochemistry. The presence of viral nucleic
acid can also be demonstrated using PCR, although since up to 30% of cats are carriers,
the amplification of viral DNA should be interpreted with caution [54]. The detection
of caliciviral nucleic acid from blood or lesions from a cat with clinical signs consistent
with this disease is good evidence to support a diagnosis of caliciviral dermatitis [74].
Virus isolation and fluorescent antibody testing are also possible [73].
Treatment
Treatment is supportive and commercially available antivirals do not inhibit replication of caliciviruses [62]. The prognosis is good for the acute nonvirulent systemic
disease cases. Controlling secondary infections, regular cleaning of discharges and
mucolytic drugs (e.g., bromhexine), or nebulization with saline are helpful. Often
cats do not eat due to the oral ulcers, and placement of a feeding tube and enteral
nutrition may be necessary in severe cases [54].
Intensive supportive treatment is recommended for cats with virulent systemic
calicivirus infection, but even with such treatments mortality rates of 30–60% have
been reported [77]. A feline calicivirus-specific antiviral phosphorodiamidate morpholino oligomer (PMO) was developed and used to treat kittens in three outbreaks
of severe calicivirus disease. In this trial, 47 of 59 kittens that were treated with the
PMO survived, but only three of 31 untreated cats survived [80]. The success of this
experimental targeted therapy suggests that newer methods of treating calicivirus
infections in cats may become available in the future.
Feline Leukemia Virus
Feline leukemia virus (FeLV) is a retrovirus that is classified within the
Gammaretrovirus genus. Infection is most commonly spread in saliva by mutual
grooming, but can also be spread by bites and through the milk. The role of FeLV
in the development of some lymphomas is well established. The role of this virus in
the development of feline skin disease is less defined, but four cutaneous manifestations of FeLV have been proposed.
Immunosuppression
As FeLV can cause significant immunodeficiency [81], it is possible that infection
could predispose to increased opportunistic skin infections. However, there is little
direct evidence supporting an increase in skin disease in cats due to FeLV infection
and cats with FeLV rarely, if ever, initially present at a veterinarian due to recurrent
or difficult to treat opportunistic skin infections [81].
Viral Diseases
379
VetBooks.ir
Giant Cell Dermatosis
This skin disease was first reported in six cats in 1994 and subsequently in an additional cat in 2005 [82, 83]. Evidence that the disease was caused by FeLV was the
consistent detection of serum FeLV antigen and the presence of FeLV proteins in the
lesions as demonstrated by immunohistochemistry. Interestingly, four of the cats had
previously been vaccinated against FeLV and the authors speculated that some vaccines could contain infectious RNA that could result in the development of giant cell
dermatosis [83]. To the authors’ knowledge, no additional cases of feline giant cell
dermatosis have been reported. This suggests that this is a rare manifestation of FeLV
infection in cats. Additionally, due to the small numbers that have been reported, it
is not possible to definitively confirm that FeLV causes giant cell dermatosis in cats.
Affected cats have been reported to present with variable clinical signs including
multiple ulcers around the head, limbs, and paws [82], patchy alopecia and scaling
starting on the dorsum, then progressing to become more generalized, or crusting
skin lesions that were predominantly confined to the head and pinnae, but can also
develop on the footpads and around the anus [83]. Pruritus was marked in many of
the cats and concurrent gingivitis was frequently detected. Cats often presented with
evidence of systemic disease, including pyrexia and anorexia.
This disease can only be diagnosed by histology. Histology of a lesion reveals
epidermal hyperplasia with the presence of prominent multinucleate keratinocytes
that can contain up to 30 nuclei. Giant cells can be present within the surface epidermis, sebaceous glands, or in the follicular infundibula [82, 83]. Disorganization
and keratinocyte atypia may be present within the affected epidermis. Inflammation
may be prominent within the underlying dermis, especially in cases in which secondary bacterial infections are present.
There are currently no treatments for this disease and all the cats that were
reported to have this disease died shortly after diagnosis [82, 83].
Feline Paw Pad Cutaneous Horns
Affected cats develop multiple horn-like lesions that typically involve multiple pads
on multiple digits (Fig. 18). While these were initially associated with FeLV [84],
subsequent studies have identified cats with horns that were not infected by FeLV
[85, 86]. Whether FeLV infection is strictly necessary for disease development is
currently uncertain.
Cats present with multiple, elongated, conical or cylindrical masses involving
the pads of multiple digits. The lesions consist almost entirely of keratin and so are
typically grey, with a hard and dry texture. Histology reveals a well-demarcated
column of dense pale orthokeratosis covering minimally to mildly hyperplastic epidermis (Fig. 19). Inflammation is typically minimal within the underlying dermis.
Treatment is surgical excision although feline paw pad, and cutaneous horns often
locally recur. As the lesions become bigger, they can develop fissures, which can
result in secondary inflammation and pain.
VetBooks.ir
380
J. S. Munday and S. Wilhelm
Fig. 18 Feline paw pad
cutaneous horn. Lesions
are grey exophytic masses
that often involve multiple
paw pads
Fig. 19 Feline paw pad
cutaneous horn. The horns
consist of a column of
dense orthokeratosis
overlying comparatively
normal epidermis (HE,
50×)
The location and appearance of these lesions typically allow clinical diagnosis
to be made, although cutaneous horns that have developed secondary to Bowenoid
in situ carcinomas or squamous cell carcinomas could require differentiation if they
develop on the paw pad.
Cutaneous Lymphoma
Associations between FeLV and cutaneous lymphoma in cats have been inconsistently detected [87–89], and it currently appears that cats with FeLV infection are,
at worst, at an only slightly increased risk of cutaneous lymphoma.
Viral Diseases
381
VetBooks.ir
Feline Infectious Peritonitis Virus
Feline coronaviral-induced vasculitis (feline infectious peritonitis, FIP) rarely affects
the skin. However, cats with FIP and skin lesions have sporadically been reported
[90–92]. In all reported cases, the skin lesions developed late in the clinical course, in
cats that also displayed more typical clinical signs such as pyrexia, lethargy, anorexia,
or ocular lesions. The involvement of multiple cutaneous blood vessels results in the
development of nonpruritic, nonpainful, raised papules over the neck and forelimbs
or more generalized over the body. Histology reveals granulomatous vasculitis and
immunohistochemistry can be used to confirm the presence of coronaviral antigens.
Feline Immunodeficiency Virus
Feline immunodeficiency virus (FIV) is a retrovirus within the Lentivirus genus. As
the virus is typically spread by fighting, it is most common in free-roaming male
cats. While experimental infection of cats can result in marked, fatal immunosuppression, natural infection of cats appears to be much less significant and the overall
life-span of FIV-infected cats does not appear to be shorter than the life-span of
uninfected cats [93].
Currently there is scant evidence that infection by FIV predisposes to feline skin
disease [94]. While some initial cases of papillomaviral (PV) skin disease of cats
were reported in cats with FIV, there have been no direct comparisons to determine
if FIV-infected cats are disproportionately affected. Additionally, rates of PV infection were not higher in FIV-infected cats than in uninfected cats [95].
FIV has been associated with a mildly increased risk of lymphoma, although
the precise role of the virus in neoplasm development is uncertain [96]. Cutaneous
lymphoma has not been associated with FIV in cats. While an association between
cutaneous SCCs and FIV infection was reported, this was suspected to be coincidental as higher rates of both SCCs and FIV infection are expected to be present in
cats that spend significant amounts of time outside [97].
References
1. Munday JS, Pasavento P. Papillomaviridae and Polyomaviridae. In: NJ ML, Dubovi EJ, editors. Fenner’s Veterinary Virology. 5th ed. London: Academic Press; 2017. p. 229–43.
2. Munday JS. Bovine and human papillomaviruses: a comparative review. Vet Pathol.
2014;51:1063–75.
3. de Villiers EM, Fauquet C, Broker TR, et al. Classification of papillomaviruses. Virology.
2004;324:17–27.
4. Tachezy R, Duson G, Rector A, et al. Cloning and genomic characterization of Felis domesticus papillomavirus type 1. Virology. 2002;301:313–21.
VetBooks.ir
382
J. S. Munday and S. Wilhelm
5. Terai M, Burk RD. Felis domesticus papillomavirus, isolated from a skin lesion, is related to
canine oral papillomavirus and contains a 1.3 kb non-coding region between the E2 and L2
open reading frames. J Gen Virol. 2002;83:2303–7.
6. Lange CE, Tobler K, Markau T, et al. Sequence and classification of FdPV2, a papillomavirus
isolated from feline Bowenoid in situ carcinomas. Vet Microbiol. 2009;137:60–5.
7. Dunowska M, Munday JS, Laurie RE, et al. Genomic characterisation of Felis catus papillomavirus 4, a novel papillomavirus detected in the oral cavity of a domestic cat. Virus Genes.
2014;48:111–9.
8. Munday JS, Dittmer KE, Thomson NA, et al. Genomic characterisation of Felis catus papillomavirus type 5 with proposed classification within a new papillomavirus genus. Vet Microbiol.
2017;207:50–5.
9. Munday JS, Dunowska M, Hills SF, et al. Genomic characterization of Felis catus papillomavirus-­3: a novel papillomavirus detected in a feline Bowenoid in situ carcinoma. Vet Microbiol.
2013;165:319–25.
10. Carney HC, England JJ, Hodgin EC, et al. Papillomavirus infection of aged Persian cats. J Vet
Diagn Investig. 1990;2:294–9.
11. Munday JS, Thomson NA, Luff JA. Papillomaviruses in dogs and cats. Vet J. 2017;225:23–31.
12. Wilhelm S, Degorce-Rubiales F, Godson D, et al. Clinical, histological and immunohistochemical study of feline viral plaques and bowenoid in situ carcinomas. Vet Dermatol.
2006;17:424–31.
13. Munday JS, Kiupel M, French AF, et al. Amplification of papillomaviral DNA sequences from
a high proportion of feline cutaneous in situ and invasive squamous cell carcinomas using a
nested polymerase chain reaction. Vet Dermatol. 2008;19:259–63.
14. Munday JS, Peters-Kennedy J. Consistent detection of Felis domesticus papillomavirus 2
DNA sequences within feline viral plaques. J Vet Diagn Investig. 2010;22:946–9.
15. Thomson NA, Dunowska M, Munday JS. The use of quantitative PCR to detect Felis catus
papillomavirus type 2 DNA from a high proportion of queens and their kittens. Vet Microbiol.
2015;175:211–7.
16. Geisseler M, Lange CE, Favrot C, et al. Geno- and seroprevalence of Felis domesticus
Papillomavirus type 2 (FdPV2) in dermatologically healthy cats. BMC Vet Res. 2016;12:147.
17. Munday JS. Papillomaviruses in felids. Vet J. 2014;199:340–7.
18. Ravens PA, Vogelnest LJ, Tong LJ, et al. Papillomavirus-associated multicentric squamous
cell carcinoma in situ in a cat: an unusually extensive and progressive case with subsequent
metastasis. Vet Dermatol. 2013;24:642–5, e161–642.
19. Sundberg JP, Van Ranst M, Montali R, et al. Feline papillomas and papillomaviruses. Vet
Pathol. 2000;37:1–10.
20. Gross TL, Ihrke PJ, Walder EJ, et al. Skin diseases of the dog and cat: clinical and histopathologic diagnosis. 2nd ed. Oxford: Blackwell Science; 2005.
21. Munday JS, Fairley R, Atkinson K. The detection of Felis catus papillomavirus 3 DNA in a
feline bowenoid in situ carcinoma with novel histologic features and benign clinical behavior.
J Vet Diagn Investig. 2016;28:612–5.
22. Munday JS, Aberdein D. Loss of retinoblastoma protein, but not p53, is associated with the
presence of papillomaviral DNA in feline viral plaques, Bowenoid in situ carcinomas, and
squamous cell carcinomas. Vet Pathol. 2012;49:538–45.
23. Munday JS, Benfell MW, French A, et al. Bowenoid in situ carcinomas in two Devon Rex cats:
evidence of unusually aggressive neoplasm behaviour in this breed and detection of papillomaviral gene expression in primary and metastatic lesions. Vet Dermatol. 2016;27:215–e255.
24. Miller RL, Gerster JF, Owens ML, et al. Imiquimod applied topically: a novel immune
response modifier and new class of drug. Int J Immunopharmacol. 1999;21:1–14.
25. Gill VL, Bergman PJ, Baer KE, et al. Use of imiquimod 5% cream (Aldara) in cats with
multicentric squamous cell carcinoma in situ: 12 cases (2002–2005). Vet Comp Oncol.
2008;6:55–64.
26. Love W, Bernhard JD, Bordeaux JS. Topical imiquimod or fluorouracil therapy for basal and
squamous cell carcinoma: a systematic review. Arch Dermatol. 2009;145:1431–8.
VetBooks.ir
Viral Diseases
383
27. Flickinger I, Gasymova E, Dietiker-Moretti S, et al. Evaluation of long-term outcome and
prognostic factors of feline squamous cell carcinomas treated with photodynamic therapy
using liposomal phosphorylated meta-tetra(hydroxylphenyl)chlorine. J Feline Med Surg
2018:1098612X17752196.
28. Munday JS, French AF, Gibson IR, et al. The presence of p16 CDKN2A protein immunostaining within feline nasal planum squamous cell carcinomas is associated with an increased
survival time and the presence of papillomaviral DNA. Vet Pathol. 2013;50:269–73.
29. Munday JS, Gibson I, French AF. Papillomaviral DNA and increased p16CDKN2A protein are
frequently present within feline cutaneous squamous cell carcinomas in ultraviolet-protected
skin. Vet Dermatol. 2011;22:360–6.
30. Altamura G, Corteggio A, Pacini L, et al. Transforming properties of Felis catus papillomavirus type 2 E6 and E7 putative oncogenes in vitro and their transcriptional activity in feline
squamous cell carcinoma in vivo. Virology. 2016;496:1–8.
31. Thomson NA, Munday JS, Dittmer KE. Frequent detection of transcriptionally active
Felis catus papillomavirus 2 in feline cutaneous squamous cell carcinomas. J Gen Virol.
2016;97:1189–97.
32. Munday JS, Knight CG, Howe L. The same papillomavirus is present in feline sarcoids from
North America and New Zealand but not in any non-sarcoid feline samples. J Vet Diagn
Investig. 2010;22:97–100.
33. Schulman FY, Krafft AE, Janczewski T. Feline cutaneous fibropapillomas: clinicopathologic
findings and association with papillomavirus infection. Vet Pathol. 2001;38:291–6.
34. Teifke JP, Kidney BA, Lohr CV, et al. Detection of papillomavirus-DNA in mesenchymal tumour cells and not in the hyperplastic epithelium of feline sarcoids. Vet Dermatol.
2003;14:47–56.
35. Munday JS, Thomson N, Dunowska M, et al. Genomic characterisation of the feline sarcoid-­
associated papillomavirus and proposed classification as Bos taurus papillomavirus type 14.
Vet Microbiol. 2015;177:289–95.
36. Munday JS, Knight CG. Amplification of feline sarcoid-associated papillomavirus DNA
sequences from bovine skin. Vet Dermatol. 2010;21:341–4.
37. Munday JS, Thomson NA, Henderson G, et al. Identification of Felis catus papillomavirus 3 in
skin neoplasms from four cats. J Vet Diagn Investig. 2018;30:324–8.
38. Munday JS, French A, Thomson N. Detection of DNA sequences from a novel papillomavirus
in a feline basal cell carcinoma. Vet Dermatol. 2017;28:236–e260.
39. Munday JS, Fairley RA, Mills H, et al. Oral papillomas associated with Felis catus papillomavirus type 1 in 2 domestic cats. Vet Pathol. 2015;52:1187–90.
40. Munday JS, Hanlon EM, Howe L, et al. Feline cutaneous viral papilloma associated with
human papillomavirus type 9. Vet Pathol. 2007;44:924–7.
41. Johnson RP, Sabine M. The isolation of herpesviruses from skin ulcers in domestic cats. Vet
Rec. 1971;89:360–2.
42. Hargis AM, Ginn PE. Feline herpesvirus 1-associated facial and nasal dermatitis and stomatitis
in domestic cats. Vet Clin North Am Small Anim Pract. 1999;29:1281–90.
43. Sanchez MD, Goldschmidt MH, Mauldin EA. Herpesvirus dermatitis in two cats without
facial lesions. Vet Dermatol. 2012;23:171–3, e135.
44. Lee M, Bosward KL, Norris JM. Immunohistological evaluation of feline herpesvirus-1 infection in feline eosinophilic dermatoses or stomatitis. J Feline Med Surg. 2010;12:72–9.
45. Suchy A, Bauder B, Gelbmann W, et al. Diagnosis of feline herpesvirus infection by immunohistochemistry, polymerase chain reaction, and in situ hybridization. J Vet Diagn Investig.
2000;12:186–91.
46. Prost C. P34 A case of exfoliative erythema multiforme associated with herpes virus 1 infection in a European cat. Vet Dermatol. 2004;15(Suppl. 1):41–69.
47. Holland JL, Outerbridge CA, Affolter VK, et al. Detection of feline herpesvirus 1 DNA in skin
biopsy specimens from cats with or without dermatitis. J Am Vet Med Assoc. 2006;229:1442–6.
48. Persico P, Roccabianca P, Corona A, et al. Detection of feline herpes virus 1 via polymerase chain reaction and immunohistochemistry in cats with ulcerative facial dermatitis,
VetBooks.ir
384
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
J. S. Munday and S. Wilhelm
eosinophilic granuloma complex reaction patterns and mosquito bite hypersensitivity. Vet
Dermatol. 2011;22:521–7.
Lamm CG, Dean SL, Estrada MM, et al. Pathology in practice. Herpesviral dermatitis. J Am
Vet Med Assoc. 2015;247:159–61.
Maggs DJ. Antiviral therapy for feline herpesvirus infections. Vet Clin North Am Small Anim
Pract. 2010;40:1055–62.
Malik R, Lessels NS, Webb S, et al. Treatment of feline herpesvirus-1 associated disease in
cats with famciclovir and related drugs. J Feline Med Surg. 2009;11:40–8.
Thomasy SM, Lim CC, Reilly CM, et al. Evaluation of orally administered famciclovir in cats
experimentally infected with feline herpesvirus type-1. Am J Vet Res. 2011;72:85–95.
Gutzwiller MER, Brachelente C, Taglinger K, et al. Feline herpes dermatitis treated with interferon omega. Vet Dermatol. 2007;18:50–4.
Nagata M, Rosenkrantz W. Cutaneous viral dermatoses in dogs and cats. Compendium.
2013;35:E1.
Bol S, Bunnik EM. Lysine supplementation is not effective for the prevention or treatment of
feline herpesvirus 1 infection in cats: a systematic review. BMC Vet Res. 2015;11:284.
Delhon GA. Poxviridae. In: NJ ML, Dubovi EJ, editors. Fenner’s Veterinary Virology. 5th ed.
London: Academic Press; 2017. p. 157–74.
Fairley RA, Mercer AA, Copland CI, et al. Persistent pseudocowpox virus infection of the skin
of a foot in a cat. NZ Vet J. 2013;61:242–3.
Fairley RA, Whelan EM, Pesavento PA, et al. Recurrent localised cutaneous parapoxvirus
infection in three cats. NZ Vet J. 2008;56:196–201.
Chantrey J, Meyer H, Baxby D, et al. Cowpox: reservoir hosts and geographic range. Epidemiol
Infect. 1999;122:455–60.
Appl C, von Bomhard W, Hanczaruk M, et al. Feline cowpoxvirus infections in Germany:
clinical and epidemiological aspects. Berl Munch Tierarztl Wochenschr. 2013;126:55–61.
McInerney J, Papasouliotis K, Simpson K, et al. Pulmonary cowpox in cats: five cases. J Feline
Med Surg. 2016;18:518–25.
Mostl K, Addie D, Belak S, et al. Cowpox virus infection in cats: ABCD guidelines on prevention and management. J Feline Med Surg. 2013;15:557–9.
Czerny CP, Wagner K, Gessler K, et al. A monoclonal blocking-ELISA for detection of orthopoxvirus antibodies in feline sera. Vet Microbiol. 1996;52:185–200.
Tryland M, Sandvik T, Holtet L, et al. Antibodies to orthopoxvirus in domestic cats in Norway.
Vet Rec. 1998;143:105–9.
Breheny CR, Fox V, Tamborini A, et al. Novel characteristics identified in two cases of feline
cowpox virus infection. JFMS Open Reports. 2017;3:2055116917717191.
Bennett M, Gaskell CJ, Baxbyt D, et al. Feline cowpox virus infection. J Small Anim Pract.
1990;31:167–73.
Godfrey DR, Blundell CJ, Essbauer S, et al. Unusual presentations of cowpox infection in cats.
J Small Animal Pract. 2004;45:202–5.
O’Halloran C, Del-Pozo J, Breheny C, et al. Unusual presentations of feline cowpox. Vet
Record. 2016;179:442–3.
Schaudien D, Meyer H, Grunwald D, et al. Concurrent infection of a cat with cowpox virus
and feline parvovirus. J Comp Pathol. 2007;137:151–4.
Jungwirth N, Puff C, Köster K, et al. Atypical cowpox virus infection in a series of cats. J
Comp Pathol. 2018;158:71–6.
Lawn R. Risk of cowpox to small animal practitioners. Vet Rec. 2010;166:631.
Czerny CP, Eis-Hubinger AM, Mayr A, et al. Animal poxviruses transmitted from cat to man:
current event with lethal end. Zentralbl Veterinarmed B. 1991;38:421–31.
Radford AD, Addie D, Belák S, et al. Feline calicivirus infection: ABCD guidelines on prevention and management. J Feline Med Surg. 2009;11:556–64.
Deschamps J-Y, Topie E, Roux F. Nosocomial feline calicivirus-associated virulent systemic
disease in a veterinary emergency and critical care unit in France. JFMS Open Reports.
2015;1:2055116915621581.
VetBooks.ir
Viral Diseases
385
75. Pedersen NC, Elliott JB, Glasgow A, et al. An isolated epizootic of hemorrhagic-like fever
in cats caused by a novel and highly virulent strain of feline calicivirus. Vet Microbiol.
2000;73:281–300.
76. Willi B, Spiri AM, Meli ML, et al. Molecular characterization and virus neutralization patterns
of severe, non-epizootic forms of feline calicivirus infections resembling virulent systemic
disease in cats in Switzerland and in Liechtenstein. Vet Microbiol. 2016;182:202–12.
77. Pesavento PA, Maclachlan NJ, Dillard-Telm L, et al. Pathologic, immunohistochemical, and
electron microscopic findings in naturally occurring virulent systemic feline calicivirus infection in cats. Vet Pathol. 2004;41:257–63.
78. Hurley KE, Pesavento PA, Pedersen NC, et al. An outbreak of virulent systemic feline calicivirus disease. J Am Vet Med Assoc. 2004;224:241–9.
79. Declercq J. Pustular calicivirus dermatitis on the abdomen of two cats following routine ovariectomy. Vet Dermatol. 2005;16:395–400.
80. Smith AW, Iversen PL, O’Hanley PD, et al. Virus-specific antiviral treatment for controlling
severe and fatal outbreaks of feline calicivirus infection. Am J Vet Res. 2008;69:23–32.
81. Hartmann K. Clinical aspects of feline retroviruses: a review. Viruses. 2012;4:2684.
82. Favrot C, Wilhelm S, Grest P, et al. Two cases of FeLV-associated dermatoses. Vet Dermatol.
2005;16:407–12.
83. Gross TL, Clark EG, Hargis AM, et al. Giant cell dermatosis in FeLV-positive cats. Vet
Dermatol. 1993;4:117–22.
84. Center SA, Scott DW, Scott FW. Multiple cutaneous horns on the footpad of a cat. Feline
Practice. 1982;12:26–30.
85. Komori S, Ishida T, Washizu M. Four cases of cutaneous horns in the foot pads of feline leukemia virus-negative cats. J Japan Vet Med Assoc. 1998;51:27–30.
86. Chaher E, Robertson E, Sparkes A, et al. Call for cases: cat paw hyperkeratosis. CVE Control
and Therapy Series. 2016;282:51–4.
87. Burr HD, Keating JH, Clifford CA, et al. Cutaneous lymphoma of the tarsus in cats: 23 cases
(2000–2012). J Am Vet Med Assoc. 2014;244:1429–34.
88. Fontaine J, Heimann M, Day MJ. Cutaneous epitheliotropic T-cell lymphoma in the cat: a
review of the literature and five new cases. Vet Dermatol. 2011;22:454–61.
89. Roccabianca P, Avallone G, Rodriguez A, et al. Cutaneous lymphoma at injection sites:
pathological, immunophenotypical, and molecular characterization in 17 cats. Vet Pathol.
2016;53:823–32.
90. Cannon MJ, Silkstone MA, Kipar AM. Cutaneous lesions associated with coronavirus-induced
vasculitis in a cat with feline infectious peritonitis and concurrent feline immunodeficiency
virus infection. J Feline Med Surg. 2005;7:233–6.
91. Martha JC, Malcolm AS, Anja MK. Cutaneous lesions associated with coronavirus-induced
vasculitis in a cat with feline infectious peritonitis and concurrent feline immunodeficiency
virus infection. J Feline Med Surg. 2005;7:233–6.
92. Bauer BS, Kerr ME, Sandmeyer LS, et al. Positive immunostaining for feline infectious peritonitis (FIP) in a Sphinx cat with cutaneous lesions and bilateral panuveitis. Vet Ophthalmol.
2013;16(Suppl 1):160–3.
93. Murphy B. Retroviridae. In: NJ ML, Dubovi EJ, editors. Fenner’s Veterinary Virology. 5th ed.
London: Academic Press; 2017. p. 269–97.
94. Backel K, Cain C. Skin as a marker of general feline health: cutaneous manifestations of infectious disease. J Feline Med Surg. 2017;19:1149–65.
95. Munday JS, Witham AI. Frequent detection of papillomavirus DNA in clinically normal
skin of cats infected and noninfected with feline immunodeficiency virus. Vet Dermatol.
2010;21:307–10.
96. Magden E, Quackenbush SL, VandeWoude S. FIV associated neoplasms—a mini-review. Vet
Immunol Immunopathol. 2011;143:227–34.
97. Hutson CA, Rideout BA, Pedersen NC. Neoplasia associated with feline immunodeficiency virus infection in cats of southern California. J Am Vet Med Assoc. 1991;199:
1357–62.
VetBooks.ir
Leishmaniosis
Maria Grazia Pennisi
Abstract
Leishmania spp. affecting cats include L. infantum, L. mexicana, L. venezuelensis, L. amazonensis, and L. braziliensis. Leishmania infantum is the species most
frequently reported in cats and causes feline leishmaniosis (FeL). Cats exposed
to L. infantum are able to mount a cell-mediated immune response that does not
parallel antibody production. Cats with L. infantum-associated clinical disease
have positive blood PCR and low to very high antibody levels. About half of the
clinical cases of FeL are diagnosed in cats with impaired immunocompetence.
Skin or mucocutaneous lesions are the most common clinical findings; however,
FeL is a systemic disease. Skin or mucocutaneous lesions and lymph node
enlargement are seen in at least half of cases, ocular or oral lesions and some
aspecific signs (weight loss, anorexia, lethargy) in about 20–30% of cases, and
many other clinical signs (e.g., respiratory, gastrointestinal) are sporadically
observed. Ulcerative and nodular lesions due to diffuse granulomatous dermatitis
are the most frequent skin manifestations, mainly distributed on the head or
­symmetrically on the distal limbs. Diagnosis can be obtained by cytology and
histology, and immunohistochemistry is useful to confirm the causative role of
Leishmania infection in the dermopathological manifestations; however, other
skin diseases may coexist with FeL. Polymerase chain reaction is used in case of
suggestive lesions with lack of parasites and for Leishmania speciation.
Comorbidities, coinfections, and chronic renal disease influence the prognosis
and should be investigated. Treatment is currently based on the same drugs used
for canine leishmaniosis, and generally clinical cure is obtained; however recurrence is possible.
M. G. Pennisi (*)
Dipartimento di Scienze Veterinarie, Università di Messina, Messina, Italy
e-mail: mariagrazia.pennisi@unime.it
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_18
387
388
M. G. Pennisi
VetBooks.ir
Introduction
Leishmaniases are protozoan diseases caused by Leishmania spp. affecting
humans and animals, but leishmaniosis is the term used for diseases in animals.
Leishmaniosis caused by Leishmania infantum is a severe, zoonotic, vector-borne
disease endemic in areas of the Old and New Worlds, with dogs as the main reservoir
[1]. In fact, the majority of infected dogs do not develop clinical signs or clinicopathological abnormalities, but they are chronically infected and infectious to sand
fly vectors. Dogs may, however, develop a mild to severe systemic disease, with
frequent skin lesions usually associated with other clinical and clinico-pathological
abnormalities. Therefore, much research interest has been focused on canine leishmaniosis (CanL), in order to prevent the infection, understand the pathomechanisms
driving infection to disease, make early and accurate diagnosis, and treat affected
dogs. Conversely, until about 25 years ago, the cat was considered a resistant host
species to Leishmania infections, based on very rare case reports, occasional post
mortem finding of the parasite in cats from endemic areas, and results from an
experimental infection study demonstrating limited infection rates [2]. Over the last
decades, an increasing number of clinical cases have been reported, and investigations with more sensitive diagnostic techniques detected a variable, but not negligible, infection rate in cats living in endemic areas. Therefore, feline leishmaniosis
(FeL) appears nowadays as an emergent disease, and the cat’s role as reservoir host
is revalued. We now know that the epidemiology of leishmaniosis is complex and
the vectorial transmission in endemic areas involves multiple host species infectious to sand flies, including the cat. Tegumentary leishmaniosis caused by dermotropic Leishmania spp. is rarely reported in both dogs and cats. Dermotropic species
infecting cats are Leishmania tropica and Leishmania major in the Old World and
Leishmania mexicana, Leishmania venezuelensis, and Leishmania braziliensis in
the Americas. Main reservoir hosts for dermotropic species are wild animals, such
as rodents.
Etiology, Diffusion, and Transmission
Leishmania genus (Kinetoplastea: Trypanosomatidae) includes diphasic and dixenous protozoans replicating as promastigotes in the gut of phlebotomine sand flies,
their natural vectors. When inoculated into vertebrate hosts by sand fly bites, promastigotes change to the non-flagellated amastigote form that multiplies by binary
fission in macrophages. Leishmania spp. detected in cats are able to infect also other
mammals (including dogs and humans) and belong to the subgenus Leishmania
(L. infantum, L. mexicana, L. venezuelensis, L. amazonensis) or Viannia (L.
braziliensis).
Leishmania infantum is the species most frequently reported in both dogs and
cats in the Old World and in Central and South America. Leishmania infantum has
been detected in cats in Mediterranean countries (Italy, Spain, Portugal, France,
Greece, Turkey, Cyprus), Iran and Brazil [3–6]. Reported antibody and blood PCR
prevalences are very variable (from nihil to >60%) and influenced by many factors
VetBooks.ir
Leishmaniosis
389
such as the local level of endemicity, selection of tested cats and analytical differences [3]. However, L. infantum antibody and molecular prevalence is usually lower
in cats compared to dogs and cases of FeL are rarer [3, 7]. Cases of both CanL and
FeL are diagnosed in non-endemic areas in dogs or cats rehomed from or travelling
to endemic areas [1, 8–13].
Sand fly transmission is the most important way of transmission of Leishmania
to humans and animals, and several studies about the feeding habit of sand flies
suggest that this is likely also in feline infection, but it has never been investigated
[3, 14–16]. Non-vectorial transmission (vertical, by blood transfusion, mating, or
bite wounds) of CanL is well known and responsible for autochthonous cases in
non-endemic areas in dogs, but we have no evidence of these ways of transmission
to and in cats [1, 10, 17, 18]. However, blood transfusion could be a source of infection in cats as proven in dogs and humans. In fact, healthy cats – similarly to healthy
dogs and humans – are found blood PCR positive in endemic areas [4–7, 19–22].
Pathogenesis
Leishmania infantum
A great number of both experimentally and field controlled prospective studies performed on CanL provided information about immunopathogenesis of CanL, but we
do not have similar studies performed on cats. In dogs, T helper 1 (Th1) immune
response responsible for protective CD4+ T cell-mediated immunity is associated
to resistance to the disease [1]. Conversely, progression of L. infantum infection
and development of lesions and clinical signs in dogs and humans are associated
with a predominant T helper 2 (Th2) immune response and the consequent nonprotective antibody production and T cell exhaustion [23]. Depending on a variable
balance between humoral and cell-mediated immunity in the infected dog, a wide
and dynamic clinical spectrum is seen in CanL, including subclinical infection, selflimiting mild disease, or severe progressive illness [1, 24]. Sick dogs with severe
clinical disease and high blood parasitemia show a high antibody level and lack in
specific IFN-γ production [25]. Similarly to what occurs in mouse experimental
models, a complex genetic background modulates the dog’s susceptibility or resistance to CanL [1, 24]. In cats, the adaptive immune response elicited by L. infantum
exposure in endemic areas was recently explored with measurement of specific antibody and IFN-γ production [26]. Some cats produced L. infantum-specific IFN-γ
and were found blood PCR negative and antibody negative or in few cases borderline positive [26]. This means that, similarly to other mammals, cats exposed to L.
infantum are able to mount a protective cell-mediated immune response that does
not parallel antibody production. The relationship between immunological pattern
and severity of disease is still unexplored in cats; however, we know that cats with L.
infantum-associated clinical disease have a high blood parasitemia and low to very
high antibody levels [3, 27–32]. Moreover, longitudinal studies found that progression of the infection toward disease is associated in cats with increasing antibody
titers, and, on the other hand, clinical improvement obtained by anti-L. infantum
VetBooks.ir
390
M. G. Pennisi
therapy is associated with a significant reduction of antibody levels, similarly to
CanL [33–36]. Coinfections with some vector-borne pathogens (e.g., Dirofilaria
immitis, Ehrlichia canis, Hepatozoon canis) can influence parasite burden and progression of CanL [37–39]. In cats, the association between retroviral, coronavirus,
Toxoplasma, or vector-borne coinfections and antibody and/or PCR positivity to L.
infantum has been explored [5, 20, 40–50]. A significant association was found only
between feline immunodeficiency virus (FIV) and L. infantum positivity in some
cases [41, 46, 48]. Moreover, more than one third of cats with FeL and tested for retroviral coinfections were found positive to FIV [a few were also positive to Feline
Leukemia Virus (FeLV)] [11, 12, 27–29, 31, 51–69]. Other FeL cases reported in
FIV and FeLV negative cats were diagnosed in animals affected by immune-mediated diseases (and treated with immunosuppressive drugs), neoplasia, or diabetes
mellitus, and we may assume that about half of the clinical cases of FeL were diagnosed in cats with impaired immunocompetence [12, 27–30, 34, 52, 59–61].
Despite the fact that skin or mucocutaneous lesions are the most common clinical
findings, FeL is considered a systemic disease as CanL. Parasites can be detected in
various other tissues, such as lymph nodes, spleen, bone marrow, eye, kidney, liver,
and gastrointestinal and respiratory tract [8].
American Dermotropic Leishmania spp.
Some scanty information about adaptive immune response of cats toward American
dermotropic Leishmania spp. can be inferred only from case reports of L. mexicana
and from an experimental infection of cats with L. braziliensis [70–72].
Delayed-type hypersensitivity skin test with L. donovani antigen was repeatedly
found negative in a cat affected by recurrent nodular dermatitis caused by L. mexicana infection, suggesting a lack of cell-mediated adaptive immune response in this
cat [70]. Anti-Leishmania antibody production seems to be limited, as of five cats
with L. mexicana tegumentary leishmaniosis only two were antibody positive at
ELISA test, although Western blot test was positive in four [71]. Moreover, in cats
intradermally infected with a human strain of L. braziliensis, a short-term antibody
production was documented after the development of skin lesions, but, frequently,
it appeared after the healing of lesions [72].
Clinical Picture
Leishmania infantum
Currently, in endemic areas FeL is far less frequently reported than CanL, but we are
probably underestimating the disease, particularly the less frequent and less severe
clinical presentations, as it occurred in the past with CanL. Furthermore, coinfections or comorbidities are frequently detected which can contribute to a clinical
misrepresentation and misdiagnosis of FeL [3, 22, 27–32]. About a hundred of clinical cases were reported over the last 30 years – mostly in Southern Europe – and
VetBooks.ir
Leishmaniosis
391
they are at present the only source of knowledge about FeL. We are therefore aware
of the current low level of evidence (III–IV) for statements and recommendations
concerning this disease.
Age range of affected cats is wide (2–21 years); however, they are mostly mature
cats (median age 7 years) at diagnosis, with very few being 2–3 years old [3, 27,
28, 32, 51, 57, 73]. Both genders are similarly represented and almost all cases are
reported in domestic short-hair cats.
Some clinical manifestations are very frequent at diagnosis – found in at least
half of the cases – such as skin or mucocutaneous lesions and lymph node enlargement. Common presentations – found in one fourth to half of the cats – are represented by ocular or oral lesions and some aspecific signs (weight loss, anorexia,
lethargy). Finally, there are many clinical signs seen in less than one fourth of the
cases. Usually affected cats display more than one clinical sign and often develop
different lesions with time.
kin and Mucocutaneous Manifestations
S
Skin or mucocutaneous manifestations were found in about two thirds of reported
cases, but they rarely were the only abnormality detected [3, 8, 27–30, 73]. In a
study from a pathology laboratory from Spain, FeL was diagnosed in 0.57% of all
skin and ocular biopsies (n = 2632) examined over a 4-year period [73].
Several dermatological entities have been described, and different presentations often coexisted or developed subsequently in the same cat. Most lesions were
observed on the head. Pruritus was rarely reported, and in most cats manifesting
pruritus, a concurrent dermatological disease was identified such as flea allergy,
eosinophilic granuloma, pemphigus foliaceus, squamous cell carcinoma (SCC), or
demodicosis [12, 67, 75, 76]. In one case, however, pruritus stopped after starting
anti-Leishmania therapy [77].
Ulcerative dermatitis is the more commonly reported skin lesion and sometimes
with a history of self-healing and recurrence of lesion. Crusty-ulcerative lesions
with raised margins were seen on pressure points (hock, carpal, and ischiatic
regions), often symmetrically, and were large up to 5 cm (Fig. 1) [27, 28, 54, 57, 64,
Fig. 1 Large ulcer with
raised margins on right
forelimb. A similar
symmetrical lesion was
present on the left forelimb
VetBooks.ir
392
M. G. Pennisi
Fig. 2 Solitary focal
ulceration on the face
(white arrow) and
conjunctival nodule
(transparent arrow)
observed in the same cat of
Fig. 1
Fig. 3 Severe facial
ulceration in a cat
diagnosed with squamous
cell carcinoma associated
with L. infantum dermal
infection
77]. Focal solitary or multiple smaller ulcers were reported on the face (Fig. 2), lips,
ears, neck, or limbs [27, 28, 34, 64, 65, 73, 77–79]. In a few cases focal or diffuse
ulcerative dermatitis affected face, trunk, or footpads [27, 63, 65, 79]. Ulceration
of the nasal planum was also reported, and in one case it was associated with concurrent SCC [30, 54, 58, 67]. Leishmania infection and SCC were found associated in biopsied tissues obtained from a deep facial ulceration (Fig. 3) in other two
cases [56, 76]. Unfortunately, the diagnosis of SCC was missed at first consultation
in two cases when only Leishmania infection was detected by cytology or histology [30, 76]. Moreover, multifocal ulcerative dermatitis caused by L. infantum was
diagnosed in a cat suffering from SCC at a different site [65]. Ulcerative dermatitis
was found associated with eosinophilic granuloma complex, and in one other case
Leishmania infection was confirmed (by serology and skin PCR) in a cat with pemphigus foliaceus [12, 73].
VetBooks.ir
Leishmaniosis
393
Nodular dermatitis is also a frequent dermatological manifestation, and single,
multiple or diffuse, firm, alopecic, non-painful nodules were detected. They are
usually small (<1 cm), mainly distributed on the head and, in descending order of
frequency, on the eyelid, ear, chin, nose, lips, and tongue [11, 27, 28, 31, 55, 64, 66,
73, 80–83]. Nodules can be found also on limbs or rarely on the trunk or the anus
[12, 55, 73]. In rare cases nodules were ulcerated [12, 66, 84].
Differently from CanL, facial or diffuse scaling and alopecia are less frequently
reported in FeL, and in few of these cases, histopathological evaluation confirmed
the presence of amastigotes in the affected skin [29, 63, 73]. Digital hyperkeratosis
was found in one case only [27].
An atypical FeL presentation that is not reported in CanL is development of hemorrhagic bullae, observed in three cases, respectively, on the nasal planum, head,
and margin of the pinna [34, 76]. However, the lesion developed on the nasal planum was histologically diagnosed as hemangioma [76]. The other two cases were
cytologically evaluated and amastigotes were found [34].
Visceral Manifestations
Lymph node enlargement is the most frequent non-dermatological finding [3]. It is
usually multicentric and can be symmetrical. Lymph nodes are firm and non-painful, and enlargement can be relevant mimicking neoplasia. Monolateral or bilateral
ocular lesions were reported in about one third of cases, but a specialistic ophthalmic examination was not performed in all cats with FeL; therefore, some less severe
ocular findings could have been missed. Conjunctivitis (including also conjunctival
nodules) and uveitis are the most frequent ocular manifestations [11, 27, 31, 34,
60, 62, 64, 68, 73]. Keratitis, keratouveitis, and chorioretinitis were diagnosed in
a few cats [27, 31, 34, 67, 78]. Panophthalmitis is the consequence of progressive
extension of diffuse granulomatous inflammation in case of late diagnosis [60, 73].
Apart from single cases of gingival ulceration, nodular glossitis, or epulid-like
lesions, chronic stomatitis and faucitis was found in about 20% of cats, and the parasite was detected in the inflamed oral tissue [27, 31–34, 52, 58, 60, 62, 66, 78, 83].
Non-specific manifestations as weight loss, anorexia, or lethargy were not very
frequent [3], and occasionally gastrointestinal (vomiting, diarrhea) or respiratory
(chronic nasal discharge, stertor, dyspnea, wheezing) signs were reported [3, 74].
Rare manifestations were icterus, fever, spleen or liver enlargement, and abortion
[3]. Interestingly, chronic leishmanial rhinitis was confirmed in some cases [58, 64,
73–75].
American Tegumentary Leishmaniosis (ATL)
A limited number of cases of feline cutaneous leishmaniosis caused by dermotropic
Leishmania species were reported in the Americas [70, 71, 85–91]. Not always
Leishmania speciation was obtained from affected cats, and L. mexicana could be
identified in nine cases [70, 71, 91], L. braziliensis in five [85–88], L. venezuelensis
in four [90], and L. amazonensis in one [89]. They were all domestic short-hair
VetBooks.ir
394
M. G. Pennisi
cats and younger (age range: 8 months–11 years; median age 4 years) than cats
with disease caused by L. infantum. The most common manifestation consisted in
solitary or multiple firm nodules as large as 3 × 2 cm. They were alopecic, variably
erythematous, or ulcerated and mainly distributed on the pinnae and the face (eyelids, nasal planum, muzzle) and rarely on the distal limbs or tail. A larger (6 cm)
interdigital ovoid lesion was reported in a cat with L. braziliensis infection [88].
Nasal or ear ulcerations were seen in two cats with L. mexicana infection and in
two others (nasal planum or medial canthus) with L. braziliensis infection [71, 86,
87]. Mucosal nodules may develop in the nasal cavity causing sneezing, stertor, and
inspiratory dyspnea [71, 85]. No other manifestations were reported in cats with
ATL; however, some followed up cases of L. venezuelensis or L. mexicana infections developed new nodular lesions at other sites [70, 90].
Diagnosis
Diagnostic testing of symptomatic cats aims to confirm Leishmania infection and
to establish a causal relationship with the clinical picture. In case of dermatological
or mucosal lesions, the cytological evaluation of impression smears from erosions
and ulcers, of scrapings from margins of deep ulcers, and of fine needle punction of
nodules can show a pyogranulomatous pattern and the presence of amastigotes (in
the cytoplasm of macrophages or extracellularly) (Fig. 4) [3, 71]. Amastigotes have
an elliptic shape with pointed ends, measure about 3–4 × 2 μm, and are characterized by the rod-shaped basophilic kinetoplast set perpendicular to the large nucleus.
Morphology of amastigotes does not allow to differentiate between Leishmania
species. In cats with leishmaniosis caused by L. infantum, amastigotes can be found
also in cytological samples from enlarged lymph nodes, bone marrow, nasal exudates, liver, and spleen and rarely in circulating neutrophils [3].
Fig. 4 Cytology from the
cutaneous lesion in Fig. 1.
Macrophagic–neutrophilic
inflammation with
numerous intracellular
(arrows) and extracellular
amastigotes. In some
extracellular amastigotes
the basophilic rod-shaped
kinetoplast is clearly
visible (arrow heads) (May
Grünwald–Giemsa stain
1000×)
VetBooks.ir
Leishmaniosis
395
Biopsy of skin or mucosal lesions is required when cytology is inconclusive
and in any case when the clinical presentation is compatible with neoplastic or
immune-mediated diseases. Amastigotes are not easily detected by the conventional
histological staining, and in suspected cases they should be investigated by immunohistochemistry (Fig. 5). However, immunohistochemistry does not allow the speciation of Leishmania amastigotes, which can be obtained by polymerase chain
reaction (PCR) and sequencing of amplicons. PCR can be performed also from
cytological slides, formalin-fixed and paraffin-embedded biopsies. Quantitative
real-time PCR is very sensitive and can provide parasite load of samples.
Dermopathological evaluation (Fig. 6) shows dermal periadnexal to diffuse
granulomatous inflammation with a diffuse infiltration of macrophages, a moderate
number of amastigotes, and a variable number of lymphocytes and plasma cells [12,
73]. The overlying epidermis is affected by hyperkeratosis, acanthosis, and ulceration [73]. In nodular lesions giants cells may be seen [73]. A low number of parasites were found in nodular lesions, characterized by perifollicular granulomatous
Fig. 5 Dark brown
amastigotes evidenced by
immunohistochemistry.
Mayer’s hematoxylin
counterstain. Bar = 10 μm.
(Courtesy of R. Puleio, IZS
Sicilia, Italy)
Fig. 6 Diffuse pyogranulomatous dermatitis (a) with numerous amastigotes within macrophages
(b). HE. Bar = 10 μm. (Courtesy of R. Puleio, IZS Sicilia, Italy)
VetBooks.ir
396
M. G. Pennisi
dermatitis and in a lichenoid interface dermatitis in a cat affected by scaly dermatitis
[73]. Mucosal (and mucocutaneous) lesions harbor a higher parasite load and submucosal diffuse granulomatous inflammation is seen [62, 68, 73]. In some cases,
a dermal, diffuse, granulomatous inflammation was found associated with lesions
characteristic of feline eosinophilic granuloma complex [54, 73]. A transepidermal
inflammatory infiltrate with parasitized macrophages was reported in the neoplastic
tissue of a cat diagnosed with concurrent SCC [56]. In another case, a stromal infiltration of parasitized macrophages was observed adjacent to islands of SCC [30].
Nodular to diffuse granulomatous dermatitis with hyperkeratotic, hyperplastic, and
often ulcerated epidermis is described in cases of ATL [71, 85, 91].
Anti-L. infantum antibody detection is performed by quantitative serology (IFAT,
ELISA, or DAT) and Western blot (WB) techniques [3]. Cutoff setting for IFAT is
established at 1:80 dilution, and almost all cats affected by clinical FeL caused by L.
infantum have low to very high antibody levels [43, 92]. Conversely, sick cats with
ATL may not have detectable circulating antibodies [71].
Culture of infected tissues provided feline strains that in most cases showed the
same zymodemes and genotypes detected in dogs or humans [3, 30].
Clinico-pathological abnormalities more frequently reported at diagnosis in cats
with FeL caused by L. infantum consisted in mild to moderate non-regenerative
anemia, hyperglobulinemia, and proteinuria [3]. Chronic kidney disease (CKD), in
most cases at an early stage (International Renal Interest Society [IRIS] stages 1 or
2), is often documented when a renal profile including urinalysis and the urine to
protein concentrations ratio is performed [32, 75].
Clinico-pathological abnormalities of cats with ATL were rarely investigated and
only eosinophilia and neutrophilia were found in one cat with L. braziliensis infection [70, 85].
Treatment and Prognosis
Treatment of cats with clinical FeL caused by L. infantum is empirical and based on
off-label use of the most common drugs prescribed to dogs with CanL [3]. Longterm oral administration of allopurinol (10–20 mg/kg once or twice daily) as monotherapy or as maintenance treatment after a course of subcutaneous injections of
meglumine antimoniate (50 mg/kg once daily for 30 days) are the most frequently
used regimens. Clinical cure is usually obtained, but efficacy and safety of used
protocols have never been evaluated in controlled studies; therefore cats should
be monitored very carefully for adverse effects during treatment (particularly cats
affected by renal disease) and for possible clinical recurrence after stopping the
therapy [3, 27–32, 34, 74]. A cutaneous adverse drug reaction (head and neck erythema, alopecia, exfoliation, and crusting) was suspected few days after starting
allopurinol in a cat [75]. The skin reaction rapidly solved after stopping allopurinol
[75]. Increases in liver enzymes were observed in another cat, and they resolved
VetBooks.ir
Leishmaniosis
397
after lowering dosage to 5 mg/kg twice a day [12]. In two further cases, acute kidney
injury was diagnosed few weeks after starting allopurinol administration [32]. In
another cat with concurrent IRIS stage 1-CKD at the time of FeL diagnosis, azotemia developed after meglumine antimoniate and afterward to miltefosine (2 mg/
kg orally once daily for 30 days) administration. [75] This latter cat was hereafter
maintained with a dietary supplementation of nucleotides and active hexose correlated compounds that was recently found effective in dogs as CanL maintenance
treatment [75, 93].
Domperidone (0.5 mg/kg orally once daily) was recently used in two cats in
association with allopurinol, and miltefosine was given in one other case [27,
29, 30].
Surgical removal of nodules was performed but generally they recurred [12, 27,
54, 81]. In one case an integrated approach between surgery and chemotherapy was
needed for treating large ulcerations [28].
Clinical recurrence is associated with raised antibody titer and parasite load [34].
Cats with clinical FeL may live for several years after diagnosis, even those
untreated and/or FIV positive, unless concurrent conditions (neoplasia) and complications (chronic kidney disease) occur or develop [32, 68].
Scant information is available about treatment and prognosis of ATL. Some cats
with L. mexicana ATL were cured after surgical excision of nodules [91]. However,
radical pinnectomy was not effective in a FIV- and FeLV-negative cat and lesions
recurred at pinnectomy site in about 2 years [70]. Subsequently new lesions progressively involved the muzzle and finally the nasal mucosa, and the cat was euthanized over 6 years after ATL diagnosis due to a mediastinal lymphosarcoma [70].
Prevention of L. infantum Infection
Individual protection of exposed cats reduces their risk to be infected by sand fly
bites and to develop the clinical disease [3, 22]. Phlebotomus perniciosus and
Lutzomyia longipalpis, proven vectors of L. infantum, respectively, in the Old and
New Worlds, were found infected after feeding on one single sick cat with FeL [33,
94]. This means that protection of cats at population level contributes to the regional
control of L. infantum infection. In fact, the percentage of antibody and/or PCRpositive cats is often not negligible in endemic areas [3–6, 20, 21, 41, 42, 45, 47].
Pyrethroids are used in dogs for preventing the bites of sand flies, but most of
them are toxic to cats [3, 95]. Collars containing a combination of 10% imidacloprid and 4.5% flumethrin are the only pyrethroid formulation licensed also for cats,
and it was effective in reducing incidence of L. infantum infection in cats living in
endemic areas [22].
According to current knowledge, testing of blood donors by antibody detection
and blood PCR is the only advisable measure for preventing non-vectorial transmission in cats [96].
398
M. G. Pennisi
VetBooks.ir
References
1. Solano-Gallego L, Koutinas A, Miró G, Cardoso L, Pennisi MG, Ferrer L, Bourdeau P, Oliva
G, Baneth G. Directions for the diagnosis, clinical staging, treatment and prevention of canine
leishmaniosis. Vet Parasitol. 2009;165:1–18.
2. Kirkpatrick CE, Farrell JP, Goldschimdt MH. Leishmania chagasi and L. donovani: experimental infections in domestic cats. Exp Parasitol. 1984;58:125–31.
3. Pennisi M-G, Cardoso L, Baneth G, Bourdeau P, Koutinas A, Miró G, Oliva G, Solano-Gallego
L. LeishVet update and recommendations on feline leishmaniosis. Parasit Vectors. 2015;8:302.
4. Can H, Döşkaya M, Özdemir HG, Şahar EA, Karakavuk M, Pektaş B, Karakuş M, Töz S, Caner
A, Döşkaya AD, İz SG, Özbel Y, Gürüz Y. Seroprevalence of Leishmania infection and molecular detection of Leishmania tropica and Leishmania infantum in stray cats of İzmir. Turkey Exp
Parasitol. 2016;167:109–14.
5. Attipa C, Papasouliotis K, Solano-Gallego L, Baneth G, Nachum-Biala Y, Sarvani E, Knowles
TG, Mengi S, Morris D, Helps C, Tasker S. Prevalence study and risk factor analysis of
selected bacterial, protozoal and viral, including vector-borne, pathogens in cats from Cyprus.
Parasit Vectors. 2017;10:130.
6. Metzdorf IP, da Costa Lima MS, de Fatima Cepa Matos M, de Souza Filho AF, de Souza
Tsujisaki RA, Franco KG, Shapiro JT, de Almeida Borges F. Molecular characterization of
Leishmania infantum in domestic cats in a region of Brazil endemic for human and canine
visceral leishmaniasis. Acta Trop. 2017;166:121–5.
7. Otranto D, Napoli E, Latrofa MS, Annoscia G, Tarallo VD, Greco G, Lorusso E, Gulotta L,
Falsone L, Basano FS, Pennisi MG, Deuster K, Capelli G, Dantas-Torres F, Brianti E. Feline
and canine leishmaniosis and other vector-borne diseases in the Aeolian Islands: Pathogen and
vector circulation in a confined environment. Vet Parasitol. 2017;236:144–15.
8. Pennisi MG. Leishmaniosis of companion animals in Europe: an update. Vet Parasitol.
2015;208:35–47.
9. Cleare E, Mason K, Mills J, Gabor M, Irwin PJ. Remaining vigilant for the exotic: cases of
imported canine leishmaniosis in Australia 2000–2011. Aust Vet J. 2014;92:119–27.
10. Svobodova V, Svoboda M, Friedlaenderova L, Drahotsky P, Bohacova E, Baneth G. Canine
leishmaniosis in three consecutive generations of dogs in Czech Republic. Vet Parasitol.
2017;237:122–4.
11. Richter M, Schaarschmidt-Kiener D, Krudewig C. Ocular signs, diagnosis and long-term treatment with allopurinol in a cat with leishmaniasis. Schweiz Arch Tierheilkd. 2014;156:289–94.
12. Rüfenacht S, Sager H, Müller N, Schaerer V, Heier A, Welle MM, Roosje PJ. Two cases of
feline leishmaniosis in Switzerland. Vet Rec. 2005;156:542–5.
13. Best MP, Ash A, Bergfeld J, Barrett J. The diagnosis and management of a case of leishmaniosis in a dog imported to Australia. Vet Parasitol. 2014;202:292–5.
14. González E, Jiménez M, Hernández S, Martín-Martín I, Molina R. Phlebotomine sand fly
survey in the focus of leishmaniasis in Madrid, Spain (2012–2014): seasonal dynamics,
Leishmania infantum infection rates and blood meal preferences. Parasit Vectors. 2017;10:368.
15. Afonso MM, Duarte R, Miranda JC, Caranha L, Rangel EF. Studies on the feeding habits of Lutzomyia (Lutzomyia) longipalpis (Lutz & Neiva, 1912) (Diptera: Psychodidae:
Phlebotominae) populations from endemic areas of American visceral leishmaniasis in
Northeastern Brazil. J Trop Med. 2012;2012:1. https://doi.org/10.1155/2012/858657.
16. Baum M, Ribeiro MC, Lorosa ES, Damasio GA, Castro EA. Eclectic feeding behavior of
Lutzomyia (Nyssomyia) intermedia (Diptera, Psychodidae, Phlebotominae) in the transmission
area of American cutaneous leishmaniasis, State of Paranà. Brazil Rev Soc Bras Med Trop.
2013;46:547–54.
17. Karkamo V, Kaistinen A, Näreaho A, Dillard K, Vainio-Siukola K, Vidgrén G, Tuoresmäki N,
Anttila M. The first report of autochthonous non-vector-borne transmission of canine leishmaniosis in the Nordic countries. Acta Vet Scand. 2014;56:84.
18. Naucke TJ, Amelung S, Lorentz S. First report of transmission of canine leishmaniosis through
bite wounds from a naturally infected dog in Germany. Parasit Vectors. 2016;9:256.
VetBooks.ir
Leishmaniosis
399
19. Pennisi MG, Hartmann K, Addie DD, Lutz H, Gruffydd-Jones T, Boucraut-Baralon C,
Egberink H, Frymus T, Horzinek MC, Hosie MJ, Lloret A, Marsilio F, Radford AD, Thiry
E, Truyen U, Möstl K. European Advisory Board on Cat Diseases. Blood transfusion in cats:
ABCD guidelines for minimising risks of infectious iatrogenic complications. J Feline Med
Surg. 2015;17:588–93.
20. Persichetti M-F, Solano-Gallego L, Serrano L, Altet L, Reale S, Masucci M, Pennisi
M-G. Detection of vector-borne pathogens in cats and their ectoparasites in southern Italy.
Parasit Vectors. 2016;9:247.
21. Akhtardanesh B, Sharifi I, Mohammadi A, Mostafavi M, Hakimmipour M, Pourafshar
NG. Feline visceral leishmaniasis in Kerman, southeast of Iran: Serological and molecular
study. J Vector Borne Dis. 2017;54:96–102.
22. Brianti E, Falsone L, Napoli E, Gaglio G, Giannetto S, Pennisi MG, Priolo V, Latrofa MS,
Tarallo VD, Solari Basano F, Nazzari R, Deuster K, Pollmeier M, Gulotta L, Colella V, DantasTorres F, Capelli G, Otranto D. Prevention of feline leishmaniosis with an imidacloprid 10%/
flumethrin 4.5% polymer matrix collar. Parasit Vectors. 2017;10:334.
23. Esch KJ, Juelsgaard R, Martinez PA, Jones DE, Petersen CA. Programmed death 1-mediated T cell exhaustion during visceral leishmaniasis impairs phagocyte function. J Immunol.
2013;191:5542–50.
24. de Vasconcelos TCB, Furtado MC, Belo VS, Morgado FN, Figueiredo FB. Canine susceptibility to visceral leishmaniasis: A systematic review upon genetic aspects, considering
breed factors and immunological concepts. Infect Genet Evol. 2019;74:103293. https://doi.
org/10.1016/j.meegid.2017.10.005.
25. Solano-Gallego L, Montserrrat-Sangrà S, Ordeix L, Martínez-Orellana P. Leishmania infantum-specific production of IFN-γ and IL-10 in stimulated blood from dogs with clinical leishmaniosis. Parasit Vectors. 2016;9:317.
26. Priolo, V, Martínez Orellana, P, Pennisi, MG, Masucci, M, Foti, M, Solano-Gallego, L.
Leishmania infantum specific production of IFNγ in stimulated blood from outdoor cats in
endemic areas. In: Proceedings World Leish 6, Toledo-Spain (16th–20th May 2017), 2017:
C1038.
27. Bardagi, M, Lloret, A, Dalmau, A, Esteban, D, Font, A, Leiva, M, Ortunez, A, Pena, T, Real,
L, Salò, F, Tabar, MD. Feline Leishmaniosis: 15 cases. In: Proceedings 8th World Congress of
Veterinary Dermatology, Toledo-Spain (31st May–4th June 2016), 2016: 112–113.
28. Basso MA, Marques C, Santos M, Duarte A, Pissarra H, Carreira LM, Gomes L, Valério-Bolas
A, Tavares L, Santos-Gomes G, Pereira da Fonseca I. Successful treatment of feline leishmaniosis using a combination of allopurinol and N-methyl-glucamine antimoniate. JFMS Open
Rep. 2016;2:205511691663000. https://doi.org/10.1177/2055116916630002.
29. Dedola, C, Ibba, F, Manca, T, Garia, C, Abramo, F. Dermatite esfoliativa associata a leishmaniosi in un gatto. Paper presented at 2° Congresso Nazionale SIDEV, Aci Castello-Catania, Italy,
17th–19th July 2015.
30. Maia C, Sousa C, Ramos C, Cristóvão JM, Faísca P, Campino L. First case of feline leishmaniosis caused by Leishmania infantum genotype E in a cat with a concurrent nasal
squamous cell carcinoma. JFMS Open Rep. 2015;1:205511691559396. https://doi.
org/10.1177/2055116915593969.
31. Pimenta P, Alves-Pimenta S, Barros J, Barbosa P, Rodrigues A, Pereira MJ, Maltez L, Gama
A, Cristóvão JM, Campino L, Maia C, Cardoso L. Feline leishmaniosis in Portugal: 3 cases
(year 2014). Vet Parasitol Reg Stud Reports. 2015;1–2:65–9. https://doi.org/10.1016/j.
vprsr.2016.02.003.
32. Pennisi MG, Persichetti MF, Migliazzo A, De Majo M, Iannelli NM, Vitale F. Feline leishmaniosis: clinical signs and course in 14 followed up cases. Atti LXX Convegno SISVET.
2016;70:166–7.
33. Maroli M, Pennisi MG, Di Muccio T, Khoury C, Gradoni L, Gramiccia M. Infection of sandflies by a cat naturally infected with Leishmania infantum. Vet Parasitol. 2007;145:357–60.
34. Pennisi MG, Venza M, Reale S, Vitale F, Lo Giudice S. Case report of leishmaniasis in four
cats. Vet Res Commun. 2004;28(Suppl 1):363–6.
VetBooks.ir
400
M. G. Pennisi
35. Foglia Manzillo V, Di Muccio T, Cappiello S, Scalone A, Paparcone R, Fiorentino E, Gizzarelli
M, Gramiccia M, Gradoni L, Oliva G. Prospective study on the incidence and progression of
clinical signs in naïve dogs naturally infected by Leishmania infantum. PLoS Negl Trop Dis.
2013;7:e2225.
36. Solano-Gallego L, Di Filippo L, Ordeix L, Planellas M, Roura X, Altet L, MartínezOrellana P, Montserrat S. Early reduction of Leishmania infantum-specific antibodies and
blood parasitemia during treatment in dogs with moderate or severe disease. Parasit Vectors.
2016;9:235.
37. De Tommasi AS, Otranto D, Dantas-Torres F, Capelli G, Breitschwerdt EB, de Caprariis
D. Are vector-borne pathogen co-infections complicating the clinical presentation in dogs?
Parasit Vectors. 2013;6:97.
38. Morgado FN, Cavalcanti ADS, de Miranda LH, O’Dwyer LH, Silva MRL, da, Menezes, R.C,
Andrade da Silva AV, Boité MC, Cupolillo E, Porrozzi R. Hepatozoon canis and Leishmania
spp. coinfection in dogs diagnosed with visceral leishmaniasis. Braz J Vet Parasitol.
2016;25:450–8.
39. Tabar MD, Altet L, Martínez V, Roura X. Wolbachia, filariae and Leishmania coinfection in
dogs from a Mediterranean area. J Small Anim Pract. 2013;54:174–8.
40. Ayllón T, Diniz PPVP, Breitschwerdt EB, Villaescusa A, Rodríguez-Franco F, Sainz A. Vectorborne diseases in client-owned and stray cats from Madrid. Spain Vector Borne Zoonotic Dis.
2012;12:143–50.
41. Pennisi MG, Masucci M, Catarsini O. Presenza di anticorpi anti-Leishmania in gatti FIV+ che
vivono in zona endemica. Atti LII Convegno SISVET. 1998;52:265–6.
42. Pennisi MG, Maxia L, Vitale F, Masucci M, Borruto G, Caracappa S. Studio sull’infezione da
Leishmania mediante PCR in gatti che vivono in zona endemica. Atti LIV Convegno SISVET.
2000;54:215–6.
43. Pennisi MG, Lupo T, Malara D, Masucci M, Migliazzo A, Lombardo G. Serological and
molecular prevalence of Leishmania infantum infection in cats from Southern Italy. J Feline
Med Surg. 2012;14:656–7.
44. Persichetti MF, Pennisi MG, Vullo A, Masucci M, Migliazzo A, Solano-Gallego L. Clinical
evaluation of outdoor cats exposed to ectoparasites and associated risk for vector-borne infections in southern Italy. Parasit Vectors. 2018;11:136.
45. Sherry K, Miró G, Trotta M, Miranda C, Montoya A, Espinosa C, Ribas F, Furlanello T,
Solano-Gallego L. A serological and molecular study of Leishmania infantum infection in cats
from the Island of Ibiza (Spain). Vector Borne Zoonotic Dis. 2011;11:239–45.
46. Sobrinho LSV, Rossi CN, Vides JP, Braga ET, Gomes AAD, de Lima VMF, Perri SHV,
Generoso D, Langoni H, Leutenegger C, Biondo AW, Laurenti MD, Marcondes M. Coinfection
of Leishmania chagasi with Toxoplasma gondii, Feline Immunodeficiency Virus (FIV) and
Feline Leukemia Virus (FeLV) in cats from an endemic area of zoonotic visceral leishmaniasis.
Vet Parasitol. 2012;187:302–6.
47. Solano-Gallego L, Rodríguez-Cortés A, Iniesta L, Quintana J, Pastor J, Espada Y, Portús
M, Alberola J. Cross-sectional serosurvey of feline leishmaniasis in ecoregions around the
Northwestern Mediterranean. Am J Trop Med Hyg. 2007;76:676–80.
48. Spada E, Proverbio D, Migliazzo A, Della Pepa A, Perego R, Bagnagatti De Giorgi G. Serological
and molecular evaluation of Leishmania infantum infection in stray cats in a nonendemic area
in Northern Italy. ISRN Parasitol. 2013;2013:1. https://doi.org/10.5402/2013/916376.
49. Spada E, Canzi I, Baggiani L, Perego R, Vitale F, Migliazzo A, Proverbio D. Prevalence
of Leishmania infantum and co-infections in stray cats in northern Italy. Comp Immunol
Microbiol Infect Dis. 2016;45:53–8.
50. Vita S, Santori D, Aguzzi I, Petrotta E, Luciani A. Feline leishmaniasis and ehrlichiosis: serological investigation in Abruzzo region. Vet Res Commun. 2005;29(Suppl 2):319–21.
51. Britti D, Vita S, Aste A, Williams DA, Boari A. Sindrome da malassorbimento in un gatto con
leishmaniosi. Atti LIX Convegno SISVET. 2005;59:281–2.
52. Caracappa S, Migliazzo A, Lupo T, Lo Dico M, Calderone S, Reale S, Currò V, Vitale
M. Analisi biomolecolari, sierologiche ed isolamento in un gatto infetto da Leishmania spp.
Atti. X Congresso Nazionale SIDiLV. 2008;10:134–5.
VetBooks.ir
Leishmaniosis
401
53. Coelho WMD, de Lima VMF, Amarante AFT, do, Langoni, H, Pereira VBR, Abdelnour A,
Bresciani KDS. Occurrence of Leishmania (Leishmania) chagasi in a domestic cat (Felis
catus) in Andradina, São Paulo, Brazil: case report. Rev Bras Parasitol Vet. 2010;19:256–8.
54. Dalmau A, Ossó M, Oliva A, Anglada L, Sarobé X, Vives E. Leishmaniosis felina a propósito
de un caso clínico. Clínica Vet Pequeños Anim. 2008;28:233–8.
55. Fileccia, I. Qual’è la vostra diagnosi? Paper presented at 1st Congresso Nazionale SIDEV,
Montesilvano (Pescara, Italia), 21–23, September 2012.
56. Grevot A, Jaussaud Hugues P, Marty P, Pratlong F, Ozon C, Haas P, Breton C, Bourdoiseau
G. Leishmaniosis due to Leishmania infantum in a FIV and FELV positive cat with a squamous
cell carcinoma diagnosed with histological, serological and isoenzymatic methods. Parasite
Paris. 2005;12:271–5.
57. Hervás J, Chacón-M De Lara F, Sánchez-Isarria MA, Pellicer S, Carrasco L, Castillo JA,
Gómez-Villamandos JC. Two cases of feline visceral and cutaneous leishmaniosis in Spain. J
Feline Med Surg. 1999;1:101–5.
58. Ibba, F. Un caso di rinite cronica in corso di leishmaniosi felina. In: Proceedings 62nd
Congresso Internazionale Multisala SCIVAC, Rimini-Italy (29th–31st May 2009), 2009:568.
59. Laruelle-Magalon C, Toga I. Un cas de leishmaniose féline. Prat Méd Chir Anim Comp.
1996;31:255–61.
60. Leiva M, Lloret A, Peña T, Roura X. Therapy of ocular and visceral leishmaniasis in a cat. Vet
Ophthalmol. 2005;8:71–5.
61. Marcos R, Santos M, Malhão F, Pereira R, Fernandes AC, Montenegro L, Roccabianca
P. Pancytopenia in a cat with visceral leishmaniasis. Vet Clin Pathol. 2009;38:201–5.
62. Migliazzo A, Vitale F, Calderone S, Puleio R, Binanti D, Abramo F. Feline leishmaniosis: a
case with a high parasitic burden. Vet Dermatol. 2015;26:69–70.
63. Ozon C, Marty P, Pratlong F, Breton C, Blein M, Lelièvre A, Haas P. Disseminated feline
leishmaniosis due to Leishmania infantum in Southern France. Vet Parasitol. 1998;75:273–7.
64. Pennisi, MG, Lupo, T, Migliazzo, A, Persichetti, M-F, Masucci, M, Vitale, F. Feline
Leishmaniosis in Italy: Restrospective evaluation of 24 clinical cases. In: Proceedings World
Leish 5, Porto de Galinhas, Pernambuco-Brazil (13th–17th May 2013), 2013:P837.
65. Pocholle E, Reyes-Gomez E, Giacomo A, Delaunay P, Hasseine L, Marty P. A case of feline
leishmaniasis in the south of France. Parasite Paris Fr. 2012;19:77–80.
66. Poli A, Abramo F, Barsotti P, Leva S, Gramiccia M, Ludovisi A, Mancianti F. Feline leishmaniosis due to Leishmania infantum in Italy. Vet Parasitol. 2002;106:181–91.
67. Sanches A, Pereira AG, Carvalho JP. Um caso de leishmaniose felina. Vet Med. 2011;63:29–30.
68. Verneuil M. Ocular leishmaniasis in a cat: case report. J Fr Ophtalmol. 2013;36:e67–72.
69. Vides JP, Schwardt TF, Sobrinho LSV, Marinho M, Laurenti MD, Biondo AW, Leutenegger
C, Marcondes M. Leishmania chagasi infection in cats with dermatologic lesions from an
endemic area of visceral leishmaniosis in Brazil. Vet Parasitol. 2011;78:22–8.
70. Barnes JC, Stanley O, Craig TM. Diffuse cutaneous leishmaniasis in a cat. J Am Vet Med
Assoc. 1993;202:416–8.
71. Rivas AK, Alcover M, Martínez-Orellana P, Montserrat-Sangrà S, Nachum-Biala Y, Bardagí
M, Fisa R, Riera C, Baneth G, Solano-Gallego L. Clinical and diagnostic aspects of feline
cutaneuous leishmaniosis in Venezuela. Parasit Vectors. 2018;11:141.
72. Simões-Mattos L, Mattos MRF, Teixeira MJ, Oliveira-Lima JW, Bevilacqua CML, Prata-Júnior
RC, Holanda CM, Rondon FCM, Bastos KMS, Coêlho ICB, Barral A, Pompeu MML. The
susceptibility of domestic cats (Felis catus) to experimental infection with Leishmania braziliensis. Vet Parasitol. 2005;127:199–208.
73. Navarro JA, Sánchez J, Peñafiel-Verdú C, Buendía AJ, Altimira J, Vilafranca
M. Histopathological lesions in 15 cats with leishmaniosis. J Comp Pathol. 2010;143:297–302.
74. Altuzarra R, Movilla R, Roura X, Espada Y, Majo N, Novella R. Computed tomography features of destructive granulomatous rhinitis with intracranial extension secondary to leishmaniasis in a cat. Vet Radiol Ultrasound. 2018; https://doi.org/10.1111/vru.12666.
75. Leal RO, Pereira H, Cartaxeiro C, Delgado E, Peleteiro MDC. Pereira da Fonseca,
I. Granulomatous rhinitis secondary to feline leishmaniosis: report of an unusual presentation
and therapeutic complications. JFMS Open Rep. 2018;4(2):2055116918811374.
VetBooks.ir
402
M. G. Pennisi
76. Laurelle-Magalon C, Toga I. Un cas de leishmaniose féline. Prat Med Chir Anim Comp.
1996;31:255–61.
77. Monteverde, V, Polizzi, D, Lupo, T, Fratello, A, Leone, C, Buffa, F, Vazzana, I, Pennisi,
MG. Descrizione di un carcinoma a cellule squamose in corso di leishmaniosi in un gatto.
Atti Congresso Nazionale ceedings of the 7th National Congress of the Italian Society of
Veterinary Laboratory Diagnostics (SiDiLV). Perugia 9–10 November. 2006;7:329–330.
78. Ennas F, Calderone S, Caprì A, Pennisi MG. Un caso di leishmaniosi felina in Sardegna.
Veterinaria. 2012;26:55–9.
79. Hervás J, Chacón-Manrique de Lara F, López J, Gómez-Villamandos JC, Guerrero MJ,
Moreno A. Granulomatous (pseudotumoral) iridociclitis associated with leishmaniasis in a
cat. Vet Rec. 2001;149:624–5.
80. Cohelo WM, Lima VM, Amarante AF, Langoni H, Pereira VB, Abdelnour A, Bresciani
KD. Occurrence of Leishmania (Leishmania) chagasi in a domestic act (Felis catus) in
Andradina, São Paulo, Brazil: case report. Rev Bras Parasitol Vet. 2010;19:256–8.
81. Costa-Durão JF, Rebelo E, Peleteiro MC, Correira JJ, Simões G. Primeiro caso de leishmaniose em gato doméstico (Felis catus domesticus) detectado em Portugal (Concelho de
Sesimbra). Nota preliminar Rev Port Cienc Vet. 1994;89:140–4.
82. Savani ES, de Oliveira Camargo MC, de Carvalho MR, Zampieri RA, dos Santos MG, D’Auria
SR, Shaw JJ, Floeter-Winter LM. The first record in the Americas of an autochtonous case of
Leishmania (Leishmania) infantum chagasi in a domestic cat (Felix catus) from Cotia County,
São Paulo State. Brazil Vet Parasitol. 2004;120:229–33.
83. Ortuñez, A, Gomez, P, Verde, MT, Mayans, L, Villa, D, Navarro, L. Lesiones granulomatosas
en la mucosa oral y lengua y multiples nodulos cutaneos en un gato causado por Leishmania
infantum. In: Proceedings Southern European Veterinary Conference (SEVC), BarcelonaSpain (30th September–3rd October 2011).
84. Attipa C, Neofytou K, Yiapanis C, Martínez-Orellana P, Baneth G, Nachum-Biala Y, BrooksBrownlie H, Solano-Gallego L, Tasker S. Follow-up monitoring in a cat with leishmaniosis
and coinfections with Hepatozoon felis and “Candidatus Mycoplasma haemominutum”.
JFMS Open Rep. 2017;3:205511691774045. https://doi.org/10.1177/2055116917740454.
85. Schubach TM, Figuereido FB, Pereira SA, Madeira MF, Santos IB, Andrade MV, Cuzzi
T, Marzochi MC, Schubach A. American cutaneous leishmaniasis in two cats from Rio de
Janeiro, Brazil: first report of natural infection with Leishmania (Viannia) braziliensis. Trans
R Soc Trop Med Hyg. 2004;98:165–7.
86. Ruiz RM, Ramírez NN, Alegre AE, Bastiani CE, De Biasio MB. Detección de Leishmania
(Viannia) braziliensis en gato doméstico de Corrientes, Argentina, por técnicas de biología
molecular. Rev Vet. 2015;26:147–50.
87. Rougeron V, Catzeflis F, Hide M, De Meeûs T, Bañuls A-L. First clinical case of cutaneous
leishmaniasis due to Leishmania (Viannia) braziliensis in a domestic cat from French Guiana.
Vet Parasitol. 2011;181:325–8.
88. Passos VM, Lasmar EB, Gontijo CM, Fernandes O, Degrave W. Natural infection of a domestic cat (Felis domesticus) with Leishmania (Viannia) in the metropolitan region of Belo
Horizonte, State of Minas Gerais. Brazil Mem Inst Oswaldo Cruz. 1996;91:19–20.
89. de Souza AI, Barros EM, Ishikawa E, Ilha IM, Marin GR, Nunes VL. Feline leishmaniasis due
to Leishmania (Leishmania) amazonensis in Mato Grosso do Sul State. Brazil Vet Parasitol.
2005;128:41–5.
90. Bonfante-Garrido R, Valdivia O, Torrealba J, García MT, Garófalo MM, Urdaneta R, Alvarado
J, Copulillo E, Momen H, Grimaldi G Jr. Cutaneous leishmaniasis in cats (Felis domesticus) caused by Leishmania (Leishmania) venezuelensis. Revista Científica, FCV-LUZ.
1996;6:187–90.
91. Trainor KE, Porter BF, Logan KS, Hoffman RJ, Snowden KF. Eight cases of feline cutaneous
leishmaniasis in Texas. Vet Pathol. 2010;47:1076–81.
VetBooks.ir
Leishmaniosis
403
92. Persichetti MF, Solano-Gallego L, Vullo A, Masucci M, Marty P, Delaunay P, Vitale F, Pennisi
MG. Diagnostic performance of ELISA, IFAT and Western blot for the detection of anti-Leishmania infantum antibodies in cats using a Bayesian analysis without a gold standard. Parasit
Vectors. 2017;10:119.
93. Segarra S, Miró G, Montoya A, Pardo-Marín L, Boqué N, Ferrer L, Cerón J. Randomized,
allopurinol-controlled trial of the effects of dietary nucleotides and active hexose correlated
compound in the treatment of canine leishmaniosis. Vet Parasitol. 2017;239:50–6.
94. da Silva SM, Rabelo PFB, Gontijo, N. de F, Ribeiro RR, Melo MN, Ribeiro VM, Michalick
MSM. First report of infection of Lutzomyia longipalpis by Leishmania (Leishmania) infantum from a naturally infected cat of Brazil. Vet Parasitol. 2010;174:150–4.
95. Brianti E, Gaglio G, Napoli E, Falsone L, Prudente C, Solari Basano F, Latrofa MS, Tarallo
VD, Dantas-Torres F, Capelli G, Stanneck D, Giannetto S, Otranto D. Efficacy of a slow-­
release imidacloprid (10%)/flumethrin (4.5%) collar for the prevention of canine leishmaniosis. Parasit Vectors. 2014;7:327.
96. Pennisi MG, Hartmann K, Addie DD, Lutz H, Gruffydd-Jones T, Boucraut-Baralon C,
Egberink H, Frymus T, Horzinek MC, Hosie MJ, Lloret A, Marsilio F, Radford AD, Thiry
E, Truyen U, Möstl K. European Advisory Board on Cat Diseases. Blood transfusion in cats:
ABCD guidelines for minimising risks of infectious iatrogenic complications. J Feline Med
Surg. 2015;17:588–93.
VetBooks.ir
Ectoparasitic Diseases
Federico Leone and Hock Siew Han
Abstract
Ectoparasitic skin diseases are extremely common in cats, and their correct identification is very important for both the cat’s and the owner’s welfare. In this chapter,
the most important feline ectoparasitic diseases will be discussed, including the
morphological features of the parasite, clinical signs, diagnostic techniques and
therapeutic options. The majority of these diseases can be diagnosed with tests that
can be easily performed during the clinical examination, such as the direct examination with a magnifying lens and microscopic examination of samples collected
with clear cell tape, by superficial and deep skin scrapings and hair plucking and
microscopic examination of ear cerumen. In some cases, the diagnostic techniques
are not particularly sensitive, and a negative result doesn’t allow ruling out the
disease: a therapeutic trial is the only way to confirm or rule out the disease. The
recent introduction on the market of new wide-spectrum parasiticidal drugs, effective to prevent flea and tick infestations and with acaricidal and insecticidal activity, will make ectoparasite control much easier. However, for many diseases there
are no registered products and standardized protocols for the feline species.
Introduction
Ectoparasitic skin diseases caused by mites and insects are very important in feline
dermatology as they are included in the differential diagnoses of many pruritic dermatological conditions. Their prevalence varies depending on the geographical area
F. Leone (*)
Clinica Veterinaria Adriatica, Senigallia (Ancona), Italy
H. S. Han
The Animal Clinic, Singapore
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_19
405
VetBooks.ir
406
F. Leone and H. S. Han
considered and on the cat’s lifestyle. Living in colonies of stray cats, breeding facilities or catteries or the potential contact with stray cats make the cat more susceptible
to parasitic infestations. Some parasitic diseases may also involve the owner and,
although these infestations are usually transient since the parasite is not adapted to
man, these zoonoses should not be underestimated.
Notoedric Mange
Notoedric mange, also known as feline scabies, is a pruritic, contagious skin disease affecting the cat, caused by the mite Notoedres cati. The mite may affect other
mammals, including man, and exceptionally the dog [1–3]. The disease prevalence
is unknown; it is thought to be rare, however epidemics are still reported in some
European countries [3, 4]. Kittens are more prone to the disease compared to adult cats.
Morphology
Notoedres cati has an oval body, ventrally flattened and dorsally convex; adult
females are approximately 225 μm long and males 150 μm long. The head carries a
short and squared rostrum. The limbs are short, with unjointed pretarsi ending with
a sucker-like structure called pulvillus, present in females only on the two front
limb pairs. The hind limbs are rudimental, do not extend beyond the mite’s body,
and carry long setae lacking suckers in both sexes (Fig. 1). The dorsal cuticle shows
fingerprint-like concentrical rings, transversal rounded scales and no spines. The
anal opening is dorsally located and the eggs are oval [4, 5].
Fig. 1 Notoedres cati,
adult mite
Ectoparasitic Diseases
407
VetBooks.ir
Life Cycle
The life cycle of Notoedres cati takes place entirely on the host (permanent parasitism). After mating on the skin surface, females burrow tunnels within the stratum corneum at a speed of 2–3 mm/day. Two to three eggs a day are laid in the
tunnels for 2–4 weeks. The six-legged larva hatches from the egg, and after two
moults as protonymph and tritonymph it becomes an adult mite. The life cycle spans
14–21 days, in favorable environmental conditions. The mite feeds on epidermal
debris and interstitial fluid.
Epidemiology
Notoedric mange is extremely contagious and transmitted by direct contact. For
this reason, cats leaving in breeding facilities, catteries or colonies are predisposed.
Where feline colonies are maintained, the disease may persist and become established; this happens commonly in urban or extra-urban areas such as cemeteries and
ruins, and in close proximity to hospitals and schools [1].
Notoedric mange is a zoonotic disease, and man can transiently be infested,
showing pruritus, papules, vesicles and crusts especially on the limbs and trunk.
In a study, 63% of people coming into contact with an infested cat showed clinical
signs of notoedric mange. Mites were detected by skin scraping in 60% of patients
examined [6]. Lesions resolve spontaneously within 3 weeks, once contact with the
infected cat is stopped [6, 7].
Clinical Signs
Initial lesions are represented by papules or crusted papules and scales, which, with
disease progression, evolve into gray-yellow thick crusts, extremely adherent to the
skin surface (Fig. 2). Lesions initially appear on the ear pinnae margins and later
involve the whole pinna, the face and the neck. With disease progression, the lesions
may become generalized. Pruritus is usually severe and self-trauma is common,
causing alopecia, erosions and ulcers and predisposing to secondary bacterial or
yeast infections [1]. Grooming and the cat’s curled up sleeping habit may cause diffusion to the limbs and perineum. If not treated, the cat may become lethargic and
dehydrated and may die in rare cases [1, 8, 9].
Diagnosis
Diagnosis requires microscopic identification of the parasite and/or its eggs and/or
its feces (round-shaped and brown) in samples collected by superficial skin scraping (Box 1). Mites are usually numerous and can be easily found, as opposite to
VetBooks.ir
408
F. Leone and H. S. Han
Fig. 2 Crusting of the
pinnae margins in a cat
with notoedric mange
Fig. 3 Superficial skin
scraping: adult mites, eggs,
and mite feces are present
Sarcoptes scabiei (Fig. 3) [1, 8]. Recently, diagnosis of notoedric mange by microscopic examination of samples collected by using clear tape has been reported, with
sensitivity comparable to skin scrapings. This technique is less traumatic and therefore indicated for difficult body sites such as lips and periocular regions [10].
Box 1: Superficial Skin Scraping: Practical Tips
• Select typical locations of the parasite (e.g., margins of ear pinnae in notoedric mange)
• If clipping is necessary, use scissors instead of clippers and leave a few
millimeters of hair, to avoid removal of material containing parasites (e.g.,
crusts)
VetBooks.ir
Ectoparasitic Diseases
409
•
•
•
•
•
Apply a few drops of mineral oil on the skin
Scrape a large area of skin superficially to avoid blood contamination
Perform multiple skin scrapings
If a large amount of material is obtained, divide it onto more slides
Mix your sample on the glass slide adding a few drops of mineral oil, if
necessary, and try to obtain a single layer
• Cover with a coverslip and observe the sample with the microscope, closing partially the diaphragm and reducing the light. This allows better visualization of the parasites
Treatment
Treatment of notoedric mange can be achieved with different acaricidal active ingredients. Registered products include a spot-on formulation containing eprinomectin,
fipronil, (S)-methoprene and praziquantel [11] and a spot-on containing moxidectin
and imidacloprid [12], which can be applied once or twice at 1-month interval.
Other protocols involving active ingredients not registered for the disease involve
the use of selamectin spot-on (6–12 mg/kg applied twice at 14 or 30 days’ interval)
[1, 13, 14], ivermectin (0.2–0.3 mg/kg subcutaneously at 14 days’ interval) [1, 7,
15] and doramectin (0.2–0.3 mg/kg subcutaneously once) [16]. The new family
of isoxazolines ectoparasiticidals has been shown to be effective in other diseases
caused by mites. There are no specific studies on feline notoedric mange, but isoxazolines are likely to be effective. Notoedric mange is extremely contagious, and all
in-contact cats must be treated to avoid re-infestations.
Otodectic Mange
Otodectic mange is a parasitic disease of the external ear canal caused by the mite
Otodectes cynotis. The mite is not species-specific and may affect cats, dogs and
other mammals. Fifty to eighty percent of cases of feline otitis externa is caused by
Otodectes cynotis, which is present all over the world [5, 17].
Morphology
Otodectes cynotis has an oval body and a long, conical rostrum. Females are 345–
451 μm long, while males are smaller (274–362 μm). The limbs are long, with short
pedicles ending with a cup-shaped, sucker-like structure used by the parasite to
move quickly within the ear cerumen. The adult mites show sexual dimorphism:
males have four pairs of long limbs, ending beyond the body, and smaller abdominal
lobes; in females, the fourth pair of legs is atrophic and does not extend beyond the
body while abdominal lobes are bigger (Fig. 4). Eggs are oval, slightly flattened on
one side and 166–206 μm long [4, 5].
410
F. Leone and H. S. Han
VetBooks.ir
Fig. 4 Otodectes cynotis,
female mite
Life Cycle
The life cycle of Otodectes cynotis takes place entirely on the host (permanent parasitism). The mite lives on the surface of the external ear canals and does not burrow.
After mating, female mites lay eggs, hatching in 4–6 days. Hexapod larvae actively
feed for 3–10 days to moult in octopod protonymphae and then deutonymphae [4,
5]. Mating, often observed during microscopic examination, involves the male mite
and the deutonympha: the male mite becomes attached to the deutonympha by using
mating suckers and, if a female develops mating occurs, while when a male mite
develops there is detachment [4, 8]. The life cycle requires 3 weeks to complete
and adult mites survive on the host for approximately 2 months. Mites feed on skin
debris and fluids stimulating production of large amounts of ear cerumen, occasionally mixed with blood [8]. Mites can survive for up to 12 days off the host, in ideal
temperature conditions [18].
Epidemiology
Otodectic mange is extremely contagious and transmission occurs primarily by
direct contact with infested cats. Common is also infestation of one ear from the
other in the same cat [19]. The disease affects kittens and adults; however, juveniles
are predisposed [19]. A temporary infestation of human beings may occur, with
papules localized predominantly on the arms and trunk [20], while parasitic otitis
is extremely rare [21].
Clinical Signs
Otodectic mange causes pruritic, erythematous and ceruminous otitis externa,
almost always bilaterally. Otodectic otitis is characterized by large amounts of
brown-black dry cerumen, resembling “coffee powder” (Fig. 5) [8]. In felines,
VetBooks.ir
Ectoparasitic Diseases
411
Fig. 5 Ear cerumen
resembling coffee grains,
typical of feline
otoacariasis
hypersensitivity to mites may occur and affected cats show severe pruritus, not
proportionate to the number of mites in the ear canal [22]. On the other hand, some
cats may have huge numbers of mites in the external ear canal without pruritus,
and this may be explained by the absence of hypersensitivity phenomena [4]. Cats
infested with Otodectes cynotis may be positive to intradermal testing for house
dust mites such as Dermatophagoides farinae, Dermatophagoides pteronyssinus
and Acarus siro [23]. Secondary bacterial or yeast infections are possible [24].
Pruritus severity is responsible for auto-traumatic lesions such as alopecia, erosions, ulcers and crusting affecting the preauricular regions, head, face and neck
and for otohematomas [17].
Extra-auricular infestation may also occur, since the mite may leave the external
ear canal and cause alopecia and miliary dermatitis in other body sites (ectopic
mites) [4, 8].
Diagnosis
Diagnosis is made by microscopic observation of the mite or its eggs (Fig. 6). The
preferred technique is microscopic examination of ear cerumen obtained with an
VetBooks.ir
412
F. Leone and H. S. Han
Fig. 6 Microscopic
examination of ear
cerumen: an egg and an
adult mite mating with a
deutonympha are
visualized
ear swab (Box 2). Samples must be obtained before applying cerumenolytic products or cleaning the ear canal. To increase the sensitivity of the test, obtaining more
samples collected from the horizontal canal by passing the swab through the otoscope cone is recommended. Mites can be visualized by otoscopic examination as
white moving dots. Superficial skin scrapings allow to detect mites in cases with
extra-auricular localization [4].
Box 2: Microscopic Examination of Ear Cerumen: Practical Tips
• Collect your sample from the ear canal with a swab
• Collect your sample before applying ceruminolytics or cleaning the ear
canal
• To collect deeper samples, use the otoscope cone to guide the swab
• Dilute the sample in mineral oil, previously applied on a glass slide
• Cover with a coverslip and observe the sample on the microscope, closing
partially the diaphragm and reducing the light. This allows better visualization of the parasites
Treatment
Many topical and systemic active ingredients are available to treat otodectic mange.
Before treatment, cleaning the ear canals with a cerumenolytic product in order to
mechanically remove the parasites and the excess of cerumen caused by the mites is
recommended [8]. Topical therapy involves acaricidals such as permethrin or thiabendazole directly applied into the ear canals. These active ingredients have limited
residual activity and require daily application for 3 weeks, to ensure that all eggs
hatch and emerging larvae are exposed to the drug, despite the fact that they are
usually registered to be used for 7–10 days [17, 25]. Otologic products not containing acaricidals are also effective, although their mechanism of action is unclear.
It is hypothesized that mites die because they cannot move and/or breathe due to
the product [26, 27]. Fipronil spot-on is not registered for otoacariasis; however, it
VetBooks.ir
Ectoparasitic Diseases
413
proved effective when one drop is applied to each ear canal and the rest between the
scapulae [28]. Systemic therapy has many advantages compared to topical therapy.
Easiness of administration increases the owner’s compliance to continuity of treatment. Systemic treatment is also effective in cases with ectopic mite localization
[17]. Among non-registered active ingredients, ivermectin administered subcutaneously at 0.2–0.3 mg/kg twice at a 14-day interval or orally once weekly for 3 weeks
has been shown to be effective [29]. Registered drugs include selamectin and moxidectin-imidacloprid spot-on, both administered twice at 1-month interval [30, 31].
An evidence-based review published in 2016 recommended the use of selamectin
or moxidectin-imidacloprid spot-on once or twice at a 30-day interval for feline
otodectic mange. There is not enough evidence to recommend other active ingredients [17].
Recently, new active ingredients belonging to the isoxazolines family have been
marketed. Sarolaner and selamectin spot-on has been registered to treat otodectic
mange and was effective with a single treatment [32]. Also as single treatments,
fluralaner alone or in association with moxidectin as spot-on was shown to be effective [33, 34]. Afoxolaner, only registered for dogs, was successfully used in cats as
single oral treatment [35]. A single application of a spot-on containing eprinomectin, fipronil, (S)-methoprene, and praziquantel was effective to prevent Otodectes
cynotis infestation in cats [36]. Regardless of the treatment chosen, all in-contact
animals must be treated due to the likelihood of contagion and presence of asymptomatic carriers [29].
Cheyletiellosis
Cheyletiellosis is a parasitic skin disease caused by mites belonging to the genus
Cheyletiella. The majority of mites of the Cheyletiellidae family are predators feeding on other mites, while some species are only ectoparasitic. The three species of
dermatological interest are Cheyletiella blakei, Cheyletiella yasguri and Cheyletiella
parasitivorax [5]. Cheyletiella shows species preference, with Cheyletiella blakei
adapted to the cat, Cheyletiella yasguri to the dog and Cheyletiella parasitivorax to
the rabbit. However, there is no strict species specificity, and interspecies infestations are possible [4, 8].
Morphology
The adult mite is large (300–500 μm long); the body is hexagonal and, according to some authors, resembles a pepper or a shield [4]. The limbs are short and
carry comb-like appendixes at the end, the rostrum is well-developed with palps
ending with two prominent curved hooks, looking like Viking horns (Fig. 7). The
three Cheyletiella species can be differentiated by observing the shape of the sensorial structure (solenidion) located on the third section of the first pair of legs. The
solenidion is heart-shaped in Cheyletiella yasguri, conical in Cheyletiella blakei
414
F. Leone and H. S. Han
VetBooks.ir
Fig. 7 Cheyletiella blakei,
adult mite
and rounded in Cheyletiella parasitivorax [4, 5]. However, species differentiation is
often difficult due to individual variation of the solenidion shape and artifacts due to
fixation for microscopy [37]. The eggs are 235–245 μm long and 115–135 μm wide
and elliptical and, unlike lice eggs, are non-operculated and loosely attached to hair
shafts by thin filaments [4, 8].
Life Cycle
The life cycle of Cheyletiella spp. takes place entirely on the host (permanent parasitism). The mite lives in the stratum corneum at the base of hair shafts, moving
quickly through the scales without burrowing and feeding on epidermal debris and
fluids. The eggs are laid along the hair shafts at 2–3 mm distance from the skin surface. The hexapod larva develops within the egg; once hatched, it moults twice as
nymphal stage and finally becomes an adult mite. The life cycle spans 14–21 days,
when environmental conditions are favorable [4, 5, 8].
Epidemiology
Cheyletiellosis is extremely contagious and transmission is usually by direct
contact [4, 8]. Less often, contagion occurs indirectly because adult female mites
can survive for up to 10 days in the environment, while immature stages and
males die quickly when off the host [4, 8]. Cheyletiella can also be carried by
fleas, lice or flies [4]. The disease occurs more commonly in young animals coming from pet shops or colonies while in adult cats can be diagnosed in debilitated
or systemically ill animals [5]. Cheyletiellosis is a zoonosis and man can be
transiently infested, showing severely pruritic macules and papules on the limbs,
VetBooks.ir
Ectoparasitic Diseases
415
trunk and buttocks [8, 38, 39]. When the affected animal is treated with an acaricidal, lesions in humans spontaneously regress within 3 weeks [8].
Clinical Signs
Clinical signs are of variable severity [8]. Most affected cats initially show exfoliative dermatitis affecting the dorso-lumbar area, with small and dry whitish scales
easily detaching from the skin surface (Fig. 8) [4, 8]. The cat’s grooming behavior
may remove both the scales and the mites and initially the disease may be slowly
progressive and remain undetected [8]. Later on, the exfoliation may become more
severe and the hair coat may look “dusty.” Many authors use the term “walking
dandruff” to describe the mites moving on the skin surface. Mites are whitish in
color and can be distinguished from scales because they move [4, 8]. Pruritus is
of variable severity, from absent to severe, and not proportionate to the number of
mites, increasing the suspicion of hypersensitivity phenomena in some cats [8, 37,
40]. Some animals present with self-traumatic lesions such as alopecia, excoriations, ulcers and crusts due to severe pruritus [8]. Miliary dermatitis or self-induced
alopecia patterns can be observed [8, 29].
Fig. 8 Severe scaling on
the dorsum of a cat with
cheyletiellosis
VetBooks.ir
416
F. Leone and H. S. Han
Fig. 9 Cheyletiella egg
attached to the hair shaft:
the egg is not operculated
Diagnosis
Diagnosis of cheyletiellosis can be made by microscopic observation of the parasite
or its eggs, although the large size of the mite sometimes allows the direct observation on the cat’s hair coat, with a magnifying lens [4, 8]. The preferred technique is
microscopic examination of samples obtained with clear cell tape (Box 3). Collection
of samples may be done directly on the cat’s fur or after combing the coat with a flea
comb. Another useful technique is superficial skin scraping, particularly if few mites
are present. Microscopic examination of hair shafts allows observation of eggs attached
to hair shafts (Fig. 9) [4, 8, 29, 40, 41]. Pruritic cats may ingest the mites or eggs
due to overgrooming and a fecal flotation test may be diagnostic [4, 8, 40]. In feces,
Cheyletiella eggs are similar to Ancylostoma eggs; however, they are three to four times
bigger (230 × 100 μm) and often embryonated [4, 8]. Identification of mites can be difficult, and in some cases the diagnosis is confirmed by a therapeutic trial [8, 40].
Box 3: Microscopic Examination of Samples Collected with Clear Tape:
Practical Tips
• Choose good-quality clear tape
• Collect your sample applying the tape to the skin multiple times (note: it is
possible that parasites are not collected, especially in long-haired cats)
• Use the coat combing technique: brush the hair coat with a flea comb or with
your hands so the sample falls on the table, which should be perfectly clean
• Collect the sample with clear tape directly from the table
• Apply a few drops of mineral oil on a glass slide and cover with the clear
tape
• Cover with a coverslip and observe the sample with the microscope, closing partially the diaphragm and reducing the light. This allows better visualization of the parasites
Ectoparasitic Diseases
417
VetBooks.ir
Treatment
There is no registered active ingredient to treat cheyletiellosis in cats. Topical (fipronil
spot-on as a single treatment) [42] or systemic (selamectin spot-on, three applications
with 1-month interval [40, 43] or ivermectin, 0.2–0.3 mg/kg subcutaneously once
every 2 weeks [41]) acaricidal products have been reported as effective.
Trombiculosis
Trombiculosis is a parasitic skin disease caused by larvae of mites belonging to the
Trombiculidae family. The disease is also called “grass itch mites” or “chiggers” in
North America, “scrub itch” in Australia and “harvest mites” in Europe [44]. Within
the Trombiculoidea superfamily, the Trombiculidae family includes approximately
1500 species, of which only approximately 50 can infest birds, mammals and man.
The most important species of veterinary interest belong to the genus Trombicula,
which groups many subgenera such as Neotrombicula and Eutrombicula. In Europe,
the most commonly involved species is Neotrombicula autumnalis, while in the
Southeastern and Central USA, Eutrombicula alfreddugesi is most often diagnosed
[5, 45]. The main feature of this family is that only the larval stage is parasitic (transient parasitism), while nymphae and adult mites are free living in the environment.
The larvae are obliged parasites, are not host-specific, and can infest many species
including man [4, 5].
Morphology
Exapod Neotrombicula autumnalis larvae are oval, 200–400 μm long and are characterized by a typical red-orange color (Fig. 10). The mouth parts include a welldeveloped rostrum and chelicerae with robust tweezer-shaped palps. The trunk
carries a pentagonal dorsal scutum (rectangular in Eutrombicula alfreddugesi) and
the body is covered by long feather-shaped setae. The limbs end with a trifurcated
claw (bifurcated in Eutrombicula alfreddugesi) used to attach to the host [4, 5].
Adult mites are non-parasitic, approximately 1 mm long and are also red-orange in
color [4].
Life Cycle
The female mite lays spherical eggs on the ground. Larvae emerge from the eggs in
a week and move actively on the ground climbing on the grass and waiting for the
host [5]. Larvae require 80% relative humidity, and for this reason they climb on
plants less than 30 cm high [45]. Once on the host, the larvae attach with the chelicera and feed through a peculiar structure called stilosoma, which is made of solidified mite saliva. This structure allows the buccal apparatus to penetrate down to the
VetBooks.ir
418
F. Leone and H. S. Han
Fig. 10 Neotrombicula
autumnalis hexapod larva;
note the bright red-orange
color
derma of the host and to feed on tissue fluids (extra-intestinal digestion) [4, 46].
During the time spent on the host, the larva grows from 0.25 mm to 0.75 mm, and
its bright red-orange color becomes pale yellow [47]. After feeding for 3–15 days,
the larvae fall on the ground to complete their life cycle in the environment. The
nymphal and adult stages are free living and mobile, and feed on small arthropods
or their eggs and fluids from plants. The life cycle spans over 50–70 days and is
strongly influenced by the season [4, 5].
Epidemiology
In Europe female mites tend to lay eggs in spring and summer and larvae are very
abundant at the end of summer and in autumn. However, depending on climate,
more than one life cycle can be completed and larvae can also be found in different
seasons [4, 48, 49].
Trombiculosis is not a zoonosis because humans get infested directly from the
environment; however, direct transmission from animals to man cannot be excluded
[47]. People working or spending time in the countryside or in forests during the larvae season are predisposed. Clinical signs are thought to be due to the irritant effect
of the mite’s saliva and to acquired hypersensitivity to salivary antigens. In nonsensitized individuals, pruritic macules and papules develop, while in sensitized
VetBooks.ir
Ectoparasitic Diseases
419
patients the pruritus is severe and associated with urticaria, papules, vesicles, fever
and enlarged lymph nodes. Lesions are mostly seen on the wrist, flexural surface
of the arm, belt line, ankle, popliteal fossa and thigh [47, 49, 50]. In children, the
“summer penile syndrome” is reported: an acute hypersensitivity reaction to mites
with erythema, edema, and pruritus to the penis and dysuria due to partial phimosis
with reduction of urinary output [51].
Neotrombicula autumnalis larvae are thought to be potential vectors of Borrelia
burgdorferi, causing Lyme disease, and Anaplasma phagocytophilum (previously
known as Ehrlichia phagocytophila), causing human granulocytic anaplasmosis, by
trans-stage or trans-ovaric transmission [52–54].
Clinical Signs
Larvae climb on plants and wait for the host, to which they attach by direct contact.
For this reason, parasites are preferentially found on body areas in contact with
the ground, such as abdomen, interdigital spaces, claw folds, muzzle and pinnae,
especially in the fold at the base of the pinnae margin (Henry’s pocket). Mites can
be visualized as red-orange aggregates (Fig. 11) [5, 48]. The facial location reflects
the first contact site of the larvae with the host, directly linked to the feline exploratory behavior, while Henry’s pocket location might be explained by the epidermal
thinness which facilitates the stilosoma formation; moreover, the pocket protects
the larvae [48].
In some cats, the infestation is completely asymptomatic, and mites can be
incidentally noticed by the owner or observed during the clinical examination for
annual vaccination [48].
Other cats show variable pruritus, from moderate to severe, possibly related to
individual hypersensitivity which may persist after the larvae abandoned the host
[5, 48]. Some cats show crusted papules and self-traumatic lesions such as alopecia,
excoriations, ulcers and crusts, depending on pruritus severity. Miliary dermatitis or
self-induced alopecia may be observed [48, 55, 56].
Fig. 11 Orange-colored
collections of parasitic
larvae can be seen to the
naked eye on the head and
pinna of a cat
VetBooks.ir
420
F. Leone and H. S. Han
Fig. 12 Superficial skin
scraping: many
Neotrombicula autumnalis
larvae
Diagnosis
Diagnosis requires a compatible history and macroscopic and microscopic observation of the parasites. Hair coat examination with a magnifying lens allows to
observe small aggregates of orange-colored larvae. Microscopic examination of
samples collected with clear cell tape or superficial skin scraping allows parasite
identification (Fig. 12) [48].
Treatment
There is currently no registered treatment for trombiculosis, and there are very
few studies on the effectiveness of acaricidals to treat this disease in cats. It is a
relatively easy disease to treat, as many ectoparasiticidal products are effective;
however, re-infestation may be common in cats with free access to infested areas.
Fipronil spray [48, 57], selamectin spot-on [48, 58], and imidacloprid-moxidectin
spot-on [48] have been successfully used with a single application. These active
ingredients seem to protect against environmental re-infestations.
Demodicosis
Feline demodicosis is an uncommon to rare parasitic skin disease caused by mites
belonging to the genus Demodex. Currently, three species have been identified
in cats, using molecular techniques: Demodex cati, Demodex gatoi and a third
unnamed species [59, 60].
Ectoparasitic Diseases
421
VetBooks.ir
Morphology
Demodex cati is very similar to Demodex canis, with minimal taxonomic differences.
The body is elongated and cigar-shaped. An adult male is 182 μm long and 20 μm
wide, while an adult female is 220 μm long and 30 μm wide [4, 61, 62]. The gnathosoma, in the frontal part of the body, is trapezoidal and carries two chelicera and two
palps. In the podosoma, the middle part of the body, there are four pairs of atrophied
limbs, each one carrying one pair of tarsal claws, distally bifurcated with a large, caudally oriented dewclaw. The terminal part of the body is the opisthosoma, accounting
for two thirds of the mite body, transversally striated and ending with a tapered point
(Fig. 13) [61]. The female reproductive system is ventrally located, below the fourth
pair of legs. In the male mite, it is in the dorsal half and corresponds to the second
pair of legs. The eggs are oval and 70.5 μm long on average [4, 61].
Demodex gatoi is smaller and stubbier and morphologically similar to Demodex
criceti, the hamster’s parasite [8, 63]. Males are 90 μm long and females are 110 μm
long [62–64]. The opisthosoma accounts for less than half of the total length of the
body, is horizontally striated and caudally rounded (Fig. 14) [63, 65]. The eggs are
oval and smaller than Demodex cati eggs [63].
Fig. 13 Demodex cati: the
opisthosoma accounts for
two thirds of the parasite
body and the tip is tapered
VetBooks.ir
422
F. Leone and H. S. Han
Fig. 14 Demodex gatoi:
the opisthosoma length is
less than half of the entire
body and the tip is rounded
The third, still unnamed Demodex species is of intermediate size, with a body
shorter and stubbier than Demodex cati but longer and more tapered than Demodex
gatoi [62, 66, 67].
Life Cycle
Demodex cati lives in the hair follicle, often located close to the exit of the sebaceous gland duct, with its head directed downward [61]. Conversely, Demodex
gatoi lives in the stratum corneum [62–64]. The environment of the third
Demodex species is unknown, since it has never been described in histopathological samples [62, 66]. Information related to the life cycle are referred only to
Demodex cati [61].
The life cycle takes place entirely on the host (permanent parasitism). Mating
occurs on the skin surface; then the fertilized female moves into the hair follicle
where it lays eggs. Six-legged larvae hatch and, after two nymphal stages, the second one moves back onto the skin surface and develops into adult, and more hair
follicles are colonized [61].
Ectoparasitic Diseases
423
VetBooks.ir
Epidemiology
The way of transmission of Demodex cati is unknown. In the dog, transmission
occurs from the mother to puppies within the first days of life, during lactation [8]. The morphologic and environmental similarities of Demodex canis and
Demodex cati suggest that the way of transmission is identical. The disease is
not contagious.
The disease caused by Demodex gatoi appears to be contagious among cats sharing the same environment, if there is enough parasitic pressure [64, 66, 68]. It is not
known if the third Demodex species is contagious. Demodex spp. are host-specific
mites and the disease is not zoonotic.
Clinical Signs
In Demodex cati demodicosis, a localized and a generalized form have been
described [4, 8]. The localized form involves the head and neck, particularly the
periorbital and perilabial regions and the chin [4, 8, 69]. The lesions are erythema,
alopecia, scales and crusts. Pruritus is variable, generally mild to absent [8, 69–71].
When the disease involves the external ear canal, it causes a bilateral ceruminous
otitis which is often reported in feline immunodeficiency virus (FIV)-positive cats
[72, 73]. A localized form has also been reported in cats affected by asthma and
chronically treated with glucocorticoids administered with aerosol [74].
The generalized form causes lesions similar to the ones observed in the localized form, but more severe and extensive, involving the muzzle, neck, trunk and
limbs or the whole body (Fig. 15) [8, 65, 69–71]. The generalized disease is often
associated with immunosuppressive therapies or concurrent systemic diseases
Fig. 15 Large alopecic
area on the dorsum of a cat
with generalized
demodicosis due to
Demodex cati
VetBooks.ir
424
F. Leone and H. S. Han
Fig. 16 Severe selfinduced lesions in a cat
with Demodex gatoi
such as diabetes mellitus, xanthomas, toxoplasmosis, systemic lupus erythematosus, hypercortisolism, retroviral infections and Bowenoid in situ carcinoma
[69, 71, 75–79]. However, in some cases an underlying disease cannot be identified [80].
In Demodex gatoi infestation, the most common clinical sign is variable
pruritus, from absent to severe, and in some cases mite hypersensitivity is suspected (Fig. 16) [8, 62, 64, 81]. Cats may show self-induced alopecia involving
the trunk, abdomen flanks or limbs or self-traumatic lesions such as alopecia,
excoriations, ulcers and crusts or papular and crusting dermatitis (miliary dermatitis) [64, 81]. This type of demodicosis is not associated with immunosuppression [81]. Infestation with different Demodex species in the same cat has
been reported [62, 65].
Diagnosis
Diagnosis of feline demodicosis is confirmed by microscopic observation of the
adult mite, its immature stages, or its eggs. Diagnostic techniques used are different depending on the involved mite species and its localization. Demodex cati lives
in the hair follicle, and the preferred diagnostic method is the deep skin scraping,
followed by microscopic examination of hair pluckings (Boxes 4 and 5) [81]. For
Demodex gatoi, a superficially located species, the suggested methods are superficial skin scrapings or microscopic examination of samples obtained by clear cell
tape [81]. These mites are small and transparent, and reducing the amount of light
going through the microscope by partially closing the diaphragm to increase contrast is advised [64, 65]. In overgrooming cats, the observation of Demodex gatoi
may be difficult, and some authors suggest a fecal examination with flotation [81,
82]. Moreover, some authors suggest to treat cats with an acaricidal whenever
Demodex gatoi is suspected [64, 81].
VetBooks.ir
Ectoparasitic Diseases
425
Box 4: Microscopic Examination of Hair Pluckings: Practical Tips
•
•
•
•
•
Carefully choose the hair shafts to examine
Use hemostats or your fingers to grab the hair base
Pluck the hair in the direction of growth
Align the hair shafts on a glass slide with a few drops of mineral oil
Cover with a coverslip and observe the sample on the microscope, closing
partially the diaphragm and reducing the light. This allows better visualization of the parasites
Box 5: Deep Skin Scrapings: Practical Tips
•
•
•
•
•
•
•
Choose your sample spot, avoiding ulcerated or fibrotic areas
Clip hair if required
Apply a few drops of mineral oil to the skin
Scrape the skin until capillary bleeding is observed
Perform multiple skin scrapings
If a large amount of material is obtained, divide it onto more slides
Mix your sample on the glass slide adding a few drops of mineral oil if
necessary and try to obtain a single layer
• Cover with a coverslip and observe the sample on the microscope, closing
partially the diaphragm and reducing the light. This allows better visualization of the parasites
Treatment
There is no registered product for feline demodicosis and there are no standardized
protocols. Various active ingredients have been used with variable results, depending on the mite species and the dosage administered. An evidence-based review
recommended the use of weekly rinses with 2% calcium sulfur [83]; however, this
product is not available in many countries.
Moderate evidence of effectiveness for both Demodex species was reported for once
or twice weekly amitraz rinses (0.0125–0.025%), which may be toxic in felines, and
for macrocyclic lactones [83]. Ivermectin may be administered both orally and subcutaneously and is effective for both species; however, failures have been reported in
Demodex gatoi cases [62, 64, 81]. Doramectin (600 μg/kg subcutaneously once weekly
for 2–3 weeks) is effective to treat Demodex cati [83, 84]. Both with ivermectin and
doramectin, severe central nervous system toxicity has been described [62].
Milbemycin oxime has been shown to be effective against Demodex cati at
1–1.5 mg/kg orally once daily for 2–7 months [74, 76], and once weekly topical imidacloprid/moxidectin for eight applications is effective against Demodex gatoi [85].
VetBooks.ir
426
F. Leone and H. S. Han
Recently, single treatment with oral fluralaner has been reported to be effective
for both Demodex species [86, 87]. Demodex gatoi is contagious and treatment of
all in-contact cats is recommended [8].
Pediculosis
Pediculosis is a lice infestation. Lice are small, wingless insects, 0.5–8 mm long,
dorso-ventrally flattened, with legs carrying strong claws to attach to the hair shafts
[4, 88]. The majority of mammals, including man and birds and excluding monotremes and bats, are infested by at least one lice species [88]. As other insects, their
body is segmented with head, thorax and abdomen; they have three pair of legs
and one pair of antennae. They spend their whole life on the host and are highly
host-specific, and many species have preferred body locations. The majority of lice
belong to the suborder Anoplura, or sucking lice, infesting only placented mammals and to suborder Ischnocera, previously called Mallophaga, biting lice infesting
mammals and birds. Sucking lice have a specialized buccal apparatus for sucking
blood, while biting lice do not feed on blood but on epidermal debris and hair [4,
88]. Felicola subrostratus is the only lice infesting cats.
Morphology
Felicola subrostratus is a biting louse (suborder Ischnocera) 1–1.5 mm long, and
its color is beige-yellowish with transverse dark bands. The head is wider than the
chest and its shape is pentagonal and frontally pointed. On the ventral surface, the
louse shows a longitudinal, median cleft adapted to the hair shaft. The antennae are
similar in both sexes and comprised of three segments. The buccal apparatus is welldeveloped and helps the lice to remain attached to the hair shaft (Fig. 17). The legs
are short, ending with a single claw [88].
Fig. 17 Felicola
subrostratus, adult louse
Ectoparasitic Diseases
427
VetBooks.ir
Life Cycle
The life cycle takes place entirely on the host (permanent parasitism), where the
female lays operculated eggs strongly attached to the hair shafts. A nymph emerges
from the egg and three moults are required to become adult. The juvenile stages are
similar to the adults, but smaller and sexually immature with undeveloped gonads
(uncomplete metamorphosis). The whole cycle requires 2–3 weeks, and a female
can lay up to 200–300 eggs in its life, which lasts for approximately 1 month [88].
Compared to other insects, lice do not have a high reproductive index; however,
females during ovodeposition produce a sticky liquid which becomes solid, cementing the egg for all its length excluding the operculus (breathing opening) to the hair
shaft. This reduces the loss of eggs and mortality of immature stages and increases
the lice population on the host [88].
Epidemiology
Lice cannot survive for more than 1–2 days off the host and generally spend all their
lives on the same host. Transmission occurs by direct contact between infested and
susceptible cats, since lice leave their host only to move to another one [88]. Being
highly host-specific, transmission occurs only among cats. In temperate climates,
seasonal fluctuation with winter increase of infestations is reported, possibly due
to the host hair coat characteristics. Long-haired cats are predisposed; however, the
most severe cases are seen in malnourished cats or cats living in poor hygienic conditions [8, 88].
Clinical Signs
Lice can infest the whole body, with preferred localization on the head, neck and
dorso-lumbar region [88]. Lesions observed on the cat vary depending on the number of parasites and severity of pruritus, which is absent to moderate [8, 88]. Some
cats are asymptomatic: lice can be observed moving on the hair shafts and often
only eggs can be seen, attached to the hair shafts and macroscopically similar to
scales. Eggs can be correctly identified by their oval shape and whitish color on
closer examination. The hair coat may appear dull, unkempt, and dirty (Fig. 18) [8].
In other cases, primary lesions such as papules and scaling may be seen, or selftraumatic secondary lesions (excoriations, crusts), self-induced alopecia or miliary
dermatitis [8].
Diagnosis
Lice and their eggs are easily identified by close observation or using a magnifying lens. Microscopic examination of hair shafts and samples collected with clear
VetBooks.ir
428
F. Leone and H. S. Han
Fig. 18 Lice infestation in
a cat: the hair coat is dull
and unkempt and looks
dirty
Fig. 19 Operculated louse
egg firmly attached to the
hair shaft
cell tape confirm the diagnosis [8]. Hair combing is also useful to collect samples
from the examination table. When no adult lice are found, but just eggs, these must
be differentiated from Cheyletiella spp. eggs, also attached to hair shafts. Lice
eggs are much bigger than Cheyletiella eggs and the operculus is dorsal (Fig. 19).
Moreover, lice eggs are attached for two thirds of their length to the hair shafts,
while Cheyletiella eggs are loosely fixed by thin fibrils.
Treatment
Lice are susceptible to the majority of insecticides in the market [8]. Currently,
registered active ingredients to treat feline pediculosis include fipronil (spot-on and
spray) [89] and selamectin spot-on [90], recently made available also in association
with sarolaner. A single treatment is recommended with all these products; however, eggs are resistant to the majority of insecticides. It is advisable repeating the
VetBooks.ir
Ectoparasitic Diseases
429
treatment after 14 days to ensure that lice emerged from eggs after the first treatment
are killed. Treatment must be extended to all in-contact cats [8, 88].
Lynxacariosis
Lynxacarus radovskyi (feline fur mites) are astigmatid mites of the Listrophoridae
family which houses small long mites specialized for grasping of the hairs of mammals. Other notable fur mites include Chirodiscoides caviae and Leporacarus gibbus
which infests the guinea pig and rabbit respectively. Lynxacarus radovskyi is characterized by a laterally compressed body, short anterior legs and has a characteristic
ability to grasp the hair shaft using a modified specialized clasping structure comprised of the propodosomal flaps and palp coxae. Its legs terminate in ambulacral discs
which are membraneous structures bearing remnants of claw that facilitates maximal
contact for hair grasping. The male of the species possesses large anal suckers used
to fasten onto the female during copulation. The female then lays eggs that hatch into
six-legged larvae and then eight-legged nymphs, which finally moult into an adult
mite (Fig. 20). Lynxacarus radovskyi feed on shed corneocytes, fungal spores, sebum
and also pollen on the host. Exact life cycle has not been fully described and transmission is through direct contact. The mite has been reported in southern parts of
the USA (Texas, Florida), Australia, New Zealand, New Caledonia, French Guyana,
Caribbean, Fiji, Malaysia, Philippines, India, Singapore and also South America, but
its incidence is thought to be under reported. Lynxacarus radovskyi is not zoonotic to
humans or any other species except Felis catus.
Clinical Signs
Most infested cats are asymptomatic, but reports of a pathological response from
susceptible host have been reported. In these cats, a self-induced, non-inflammatory, caudally directed alopecia has been described. Alopecia typically begins from
Fig. 20 A typical hair
pluck demonstrating a
female nymph of
Lynxacarus radovskyi and
an egg
VetBooks.ir
430
F. Leone and H. S. Han
Fig. 21 A cat with
lynxacariosis, presented
with bilaterally
symmetrical, noninflammatory, self-­induced
alopecia
the perineum/tail base where the mites are thought to be most commonly isolated
before spreading to the lateral thighs, abdomen, and flanks (Fig. 21) [91]. Infested
cats are often presented with increased scale production and a dry, dull coat with
easily epilated hairs. Other extracutaneous signs such as gingivitis, gastrointestinal
disturbances (hairballs) and restlessness due to irritation may also be seen. As the
mite can cause extracutaneous signs, it is important that attending veterinarians consider this parasite as a possible differential diagnosis, especially when these extracutaneous signs are present.
Diagnosis
The parasites can be detected via microscopic examination of hair plucks or adhesive
tape technique obtained from the perineum, lateral hind limbs or cervical region where
they are more readily observed [91]. Mites are easily demonstrated in overt heavy
infestation but may be difficult to demonstrate in a patient that excessively self-grooms.
Treatment
The parasite is sensitive to all acaricidals. Published efficacy reports include fipronil,
moxidectin plus imidacloprid and fluralaner [92, 93]. Topical selamectin, administered every fortnightly, is equally effective.
Feline Cutaneous Screwworm Myiasis
Myiasis is defined as the invasion of a living vertebrate animal by fly larvae, which
may or may not be associated with feeding on the host tissue [94]. In cases of
obligatory myiasis, fly species such as New-World screwworm (NWS) Cochliomyia
hominivorax or the Old-World screwworm (OWS) Chrysomya bezziana lay their
eggs on a living host regardless of species, and the disease they cause is referred to
as cutaneous screwworm myiasis. Historically, the range of NWS extended from
Ectoparasitic Diseases
b
VetBooks.ir
a
431
Fig. 22 (a) A cat presented with an exudative, ulcerative, swollen and erythematous wound at the
base of the left ear, with characteristic putrid smell. (b) Upon closer inspection, burrowing larvae
are clearly visualized within these lesions
the southern states of the USA, through Mexico, Central america, the Caribbean
and northern countries of South America to Uruguay, northern Chile and northern
Argentina. Its distribution contracts during the winter months and expanded during
summer months, thus producing a seasonality at its edges and year round incidence
in the central areas. With the successful implementation of sterile insect technique (SIT), NWS has been eradicted from USA, Mexico, Curacao, Puerto Rico,
extending to Central American countries such as Guatemala, Belize, El Salvador,
Honduras, Nicaragua to Panama. OWS, as the name suggest is confined to the Old
World which includes much of Africa (from Ethiopia and sub-Saharan countries to
northern South Africa), Middle East Gulf region, the Indian subcontinent and south
east Asia (Malaysia, Singapore, Indonesia, Philipines to Papua New Guinea). OWS
has recently been reported in Hong Kong and the southern autonomous region of
Guangxi, in mainland China [95].
Clinical Signs
Female flies of Cochliomyia hominivorax and Chrysomya bezziana typically lay
their eggs at wound edges. Eggs hatch within 12–24 hours. As the name screwworm
suggests, the hatched larvae burrow or screw themselves head facing downward
into host tissue and begin feeding. This results in exudative and ulcerative lesions
with easily visualized maggots within the lesions, emanating a characteristic putrid
odor (Figs. 22a, b). These foul putrid wounds then attract more oviposition, resulting in superinfestation that can lead to death due to sepsis in the untreated host.
After approximately 7 days of feeding, the larvae drop onto the ground, burrow and
pupate, and adult flies emerge from the puparium in approximately 7 days. Adult,
intact male domestic short hair cats are predisposed to develop screwworm myiasis
VetBooks.ir
432
F. Leone and H. S. Han
from inter-cat aggression, with some concurrently diagnosed with sporotrichosis in
regions where these two diseases are reported. The most common sites for screwworm myiasis in the cats are the paws, followed by the tail and perineum [96].
Judging from the severity of tissue destruction, one would reasonably expect that the
host-parasite relationship will be highlighted by the quick removal of these larvae by
the fastidiously grooming host. However, the ability of the larvae to induce a state
of immune supression renders the host extremely tolerant of the infestation and thus
some patients are presented for medical attention in advanced stages of infestation.
Treatment
Nitenpyram (Capstar®, Elanco, IL, USA) at standard packaging dose, administered as
recommended by manufacturer (with or without food), is the most common treatment
modality in countries where the drug is available. Larvicidal efficacy is thought to
range between 94.1% to 100% within 24 hours in dogs treated with nitenpyram with
scarce data from cats [97–99]. Once the larvae have died, they are manually removed,
the wound is debrided, and if no larvae are left inside the wound (foreign body), the
wound typically heals quickly. In regions where nitenpyram is not available, extralabel use of systemic/topical ivermectin (0.3–0.6 mg/kg) and/or topical powder-based
insecticides marketed for the treatment of cutaneous myiasis in farm animals, consisting of coumaphos, propoxur, and sulphanilamide (Negasunt™ Dusting Powder,
Bayer Pharmaceuticals, Maharashtra, India), are used. There are very limited treatment options available for veterinarians to treat feline cutaneous myiasis other than
nitenpyram. Due to this limitation, many cats are still treated with extra-label use of
ivermectin and carbamates, originally meant for farm use.
References
1. Leone F, Albanese F, Fileccia I. Feline notoedric mange: a report of 22 cases. Prat Méd Chir
Anim Comp. 2003;38:421–7.
2. Leone F. Canine notoedric mange. Vet Dermatol. 2007;18(2):127–9.
3. Foley J, Serieys LE, Stephenson N, et al. A synthetic review of notoedres species mites and
mange. Parasitology. 2016;9:1–15.
4. Bowman DD, Hendrix CM, Lindsay, et al. The Arthropods. In: Bowman D, editor. Feline
Clinical Parasitology. Ames: Iowa State University Press; 2002. p. 355–455.
5. Wall R, Shearer D. Mites (Acari). In: Veterinary Ectoparasites: biology, pathology & control.
2nd ed. Oxford: Blackwell Science; 2001. p. 23–54.
6. Chakrabarti A. Human notoedric scabies from contact with cats infested with Notoedres cati.
Int J Dermatol. 1986;25(10):646–8.
7. Foley RH. A notoedric mange epizootic in an island’s cat population. Feline Pract.
1991;19:8–10.
8. Miller WH, Griffin CE, Campbell KL. Parasitic skin disease. In: Muller and Kirk’s small animal dermatology. 7th ed. St. Louis: Elsevier Mosby; 2013. p. 284–342.
9. Leone F, Albanese F, Fileccia I. Epidemiological and clinical finding of notoedric mange in 30
cats. Vet Dermatol. 2005;16(5):359.
10. Sampaio KO, de Oliveira LM, Burmann PM, et al. Acetate tape impression test for diagnosis
of notoedric mange in cats. J Feline Med Surg. 2016;15:1–4.
11. Knaus M, Capári B, Visser M. Therapeutic efficacy of Broadline against notoedric mange in
cats. Parasitol Res. 2014;113(11):4303–6.
VetBooks.ir
Ectoparasitic Diseases
433
12. Hellmann K, Petry G, Capari B, et al. Treatment of naturally Notoedres cati-infested cats with
a combination of imidacloprid 10%/moxidectin 1% spot-on (advocate®/advantage® multi,
Bayer). Parasitol Res. 2013;112(Suppl 1):57–66.
13. Itoh N, Muraoka N, Aoki M, et al. Treatment of Notoedric cati infestation in cats with selamectin. Vet Rec. 2004;154(13):409.
14. Fisher MA, Shanks DJ. A review of the off-label use of selamectin (stronghold/revolution) in
dogs and cats. Acta Vet Scand. 2008;50:46.
15. Sivajothi S, Sudhakara Reddy B, et al. Notoedres cati in cats and its management. J Parasit Dis.
2015;39(2):303–5.
16. Delucchi L, Castro E. Use of doramectin for treatment of notoedric mange in five cats. J Am
Vet Med Assoc. 2000;216(2):215–6.
17. Yang C, Huang HP. Evidence-based veterinary dermatology: a review of published studies of treatments for Otodectes cynotis (ear mite) infestation in cats. Vet Dermatol.
2016;27(4):221–e56.
18. Otranto D, Milillo P, Mesto P, et al. Otodectes cynotis (Acari: Psoroptidae): examination of survival off-the-host under natural and laboratory conditions. Exp Appl Acarol.
2004;32(3):171–9.
19. Sotiraki ST, Koutinas AF, Leontides LS, et al. Factors affecting the frequency of ear canal and
face infestation by Otodectes cynotis in the cat. Vet Parasitol. 2001;96(4):309–15.
20. Herwick RP. Lesions caused by canine ear mites. Arch Dermatol. 1978;114(1):130.
21. Lopez RA. Of mites and man. J Am Vet Med Assoc. 1993;203(5):606–7.
22. Powell MB, et al. Reaginic hypersensitivity in Otodectes cynotis infestation of cats and mode
of mite feeding. Am J Vet Res. 1980;41(6):877–82.
23. Saridomichelakis MN, Koutinas AF, Gioulekas D, et al. Sensitization to dust mites in cats with
Otodectes cynotis infestation. Vet Dermatol. 1999;10(2):89–94.
24. Roy J, Bédard C, Moreau M, et al. Comparative short-term efficacy of Oridermyl(®) auricular
ointment and revolution(®) selamectin spot-on against feline Otodectes cynotis and its associated secondary otitis externa. Can Vet J. 2012;53(7):762–6.
25. Ghubash R. Parasitic miticidal therapy. Clin Tech Small Anim Pract. 2006;21(3):135–44.
26. Scherk-Nixon M, Baker B, Pauling GE, et al. Treatment of feline otoacariasis with 2 otic
preparations not containing miticidal active ingredients. Can Vet J. 1997;38(4):229–30.
27. Engelen MA, Anthonissens E. Efficacy of non-acaricidal containing otic preparations in the
treatment of otoacariasis in dogs and cats. Vet Rec. 2000;147(20):567–9.
28. Coleman GT, Atwell RB. Use of fipronil to treat ear mites in cats. Aust Vet Pract.
1999;29(4):166–8.
29. Curtis CF. Current trends in the treatment of Sarcoptes, Cheyletiella and Otodectes mite infestations in dogs and cats. Vet Dermatol. 2004;15(2):108–14.
30. Shanks DJ, McTier TL, Rowan TG, et al. The efficacy of selamectin in the treatment of
naturally acquired aural infestations of otodectes cynotis on dogs and cats. Vet Parasitol.
2000;91(3–4):283–90.
31. Fourie LJ, Kok DJ, Heine J. Evaluation of the efficacy of an imidacloprid 10%/moxidectin 1%
spot-on against Otodectes cynotis in cats. Parasitol Res. 2003;90(Suppl 3):S112–3.
32. Becskei C, Reinemeyer C, King VL, et al. Efficacy of a new spot-on formulation of selamectin
plus sarolaner in the treatment of Otodectes cynotis in cats. Vet Parasitol. 2017;238(Suppl
1):S27–30.
33. Taenzler J, de Vos C, Roepke RK, et al. Efficacy of fluralaner against Otodectes cynotis infestations in dogs and cats. Parasit Vectors. 2017;10(1):30.
34. Taenzler J, de Vos C, Roepke RKA, et al. Efficacy of fluralaner plus moxidectin (Bravecto®
plus spot-on solution for cats) against Otodectes cynotis infestations in cats. Parasit Vectors.
2018;11(1):595.
35. Machado MA, Campos DR, Lopes NL, et al. Efficacy of afoxolaner in the treatment of otodectic mange in naturally infested cats. Vet Parasitol. 2018;256:29–31.
36. Beugnet F, Bouhsira E, Halos L, et al. Preventive efficacy of a topical combination of fipronil –
(S)-methoprene – eprinomectin – praziquantel against ear mite (Otodectes cynotis) infestation
of cats through a natural infestation model. Parasite. 2014;21:40.
37. Schmeitzel LP. Cheyletiellosis and scabies. Vet Clin North Am Small Anim Pract.
1988;18(5):1069–76.
VetBooks.ir
434
F. Leone and H. S. Han
38. Lee BW. Cheyletiella dermatitis: a report of fourteen cases. Cutis. 1991;47(2):111–4.
39. Wagner R, Stallmeister N. Cheyletiella dermatitis in humans, dogs and cats. Br J Dermatol.
2000;143(5):1110–2.
40. Chailleux N, Paradis M. Efficacy of selamectin in the treatment of naturally acquired cheyletiellosis in cats. Can Vet J. 2002;43(10):767–70.
41. Paradis M, Scott D, Villeneuve A. Efficacy of ivermectin against Cheyletiella blakei infestation
in cats. J Am Anim Hosp Assoc. 1990;26(2):125–8.
42. Scarampella F, Pollmeier M, Visser M, et al. Efficacy of fipronil in the treatment of feline
cheyletiellosis. Vet Parasitol. 2005;129(3–4):333–9.
43. Fisher MA, Shanks DJ. A review of the off-label use of selamectin (stronghold/revolution) in
dogs and cats. Acta Vet Scand. 2008;50:46.
44. Takahashi M, Misumi H, Urakami H, et al. Trombidiosis in cats caused by the bite of the larval
trombiculid mite Helenicula miyagawai (Acari: Trombiculidae). Vet Rec. 2004;154(15):471.
45. McClain D, Dana AN, Goldenberg G. Mite infestations. Dermatol Ther. 2009;22(4):327–46.
46. Shatrov AB. Stylostome formation in trombiculid mites (Acariformes: Trombiculidae). Exp
Appl Acarol. 2009;49(4):261–80.
47. Caputo V, Santi F, Cascio A, et al. Trombiculiasis: an underreported ectoparasitosis in Sicily.
Infez Med. 2018;26(1):77–80.
48. Leone F, Di Bella A, Vercelli A, et al. Feline trombiculosis: a retrospective study in 72 cats. Vet
Dermatol. 2013;24(5):535–e126.
49. Guarneri C, Chokoeva AA, Wollina U, et al. Trombiculiasis: not only a matter of animals!
Wien Med Wochenschr. 2017;167(3–4):70–3.
50. Guarneri F, Pugliese A, Giudice E, et al. Trombiculiasis: clinical contribution. Eur J Dermatol.
2005;15(6):495–6.
51. Smith GA, Sharma V, Knapp JF, et al. The summer penile syndrome: seasonal acute hypersensitivity reaction caused by chigger bites on the penis. Pediatr Emerg Care. 1998;14(2):116–8.
52. Fernández-Soto P, Pérez-Sánchez R, Encinas-Grandes A. Molecular detection of Ehrlichia
phagocytophila genogroup organisms in larvae of Neotrombicula autumnalis (Acari:
Trombiculidae) captured in Spain. J Parasitol. 2001;87(6):1482–3.
53. Kampen H, Schöler A, Metzen M, et al. Neotrombicula autumnalis (Acari, Trombiculidae) as
a vector for Borrelia burgdorferi sensu lato? Exp Appl Acarol. 2004;33(1–2):93–102.
54. Literak I, Stekolnikov AA, Sychra O, et al. Larvae of chigger mites Neotrombicula spp. (Acari:
Trombiculidae) exhibited Borrelia but no Anaplasma infections: a field study including birds
from the Czech Carpathians as hosts of chiggers. Exp Appl Acarol. 2008;44(4):307–14.
55. Leone F, Cornegliani L, Vercelli A. Clinical findings of trombiculiasis in 50 cats. Vet Dermatol.
2010;21(5):538.
56. Fleming EJ, Chastain CB. Miliary dermatitis associated with Eutrombicula infestation in a cat.
J Am Anim Hosp Assoc. 1991;27:529–31.
57. Nuttall TJ, French AT, Cheetham HC, et al. Treatment of Trombicula autumnalis infestation in
dogs and cats with a 0.25 per cent fipronil pump spray. J Small Anim Pract. 1998;39(5):237–9.
58. Leone F, Albanese F. Efficacy of selamectin spot-on formulation against Neotrombicula
autumnalis in eight cats. Vet Dermatol. 2004;15(Suppl.1):49.
59. Frank LA, Kania SA, Chung K, et al. A molecular technique for the detection and differentiation of Demodex mites on cats. Vet Dermatol. 2013;24(3):367–9. e82–e3
60. Ferreira D, Sastre N, Ravera I, et al. Identification of a third feline Demodex species through
partial sequencing of the 16S rDNA and frequency of Demodex species in 74 cats using a PCR
assay. Vet Dermatol. 2015;26(4):239–e53.
61. Desch C, Nutting WB. Demodex cati Hirst 1919: a redescription. Cornell Vet. 1979;69(3):280–5.
62. Löwenstein C, Beck W, Bessmann K, et al. Feline demodicosis caused by concurrent infestation with Demodex cati and an unnamed species of mite. Vet Rec. 2005;157(10):290–2.
63. Desch CE Jr, Stewart TB. Demodex gatoi: new species of hair follicle mite (Acari: Demodecidae)
from the domestic cat (Carnivora: Felidae). J Med Entomol. 1999;36(2):167–70.
64. Saari SA, Juuti KH, Palojärvi JH, et al. Demodex gatoi-associated contagious pruritic dermatosis in cats – a report from six households in Finland. Acta Vet Scand. 2009;51:40.
65. Neel JA, Tarigo J, Tater KC, et al. Deep and superficial skin scrapings from a feline immunodeficiency virus-positive cat. Vet Clin Pathol. 2007;36(1):101–4.
VetBooks.ir
Ectoparasitic Diseases
435
66. Kano R, Hyuga A, Matsumoto J, et al. Feline demodicosis caused by an unnamed species. Res
Vet Sci. 2012;92(2):257–8.
67. Moriello KA, Newbury S, Steinberg H. Five observations of a third morphologically distinct
feline Demodex mite. Vet Dermatol. 2013;24(4):460–2.
68. Morris DO. Contagious demodicosis in three cats residing in a common household. J Am
Anim Hosp Assoc. 1996;32(4):350–2.
69. Guaguere E, Muller A, Degorce-Rubiales F. Feline demodicosis: a retrospective study of 12
cases. Vet Dermatol. 2004;15(Suppl 1):34.
70. Stogdale L, Moore DJ. Feline demodicosis. J Am Anim Hosp Assoc. 1982;18:427–32.
71. Medleau L, Brown CA, Brown SA, et al. Demodicosis in cats. J Am Anim Hosp Assoc.
1988;24:85–91.
72. Kontos V, Sotiraki S, Himonas C. Two rare disorders in the cat: Demodectic otitis externa and Sarcoptic mange. Feline Pract. 1998;26(6):18–20.
73. Van Poucke S. Ceruminous otitis externa due to Demodex cati in a cat. Vet Rec.
2001;149(21):651–2.
74. Bizikova P. Localized demodicosis due to Demodex cati on the muzzle of two cats treated with
inhalant glucocorticoids. Vet Dermatol. 2014;25(3):222–5.
75. White SD, Carpenter JL, Moore FM, et al. Generalized demodicosis associated with diabetes
mellitus in two cats. J Am Vet Med Assoc. 1987;191(4):448–50.
76. Vogelnest LJ. Cutaneous xanthomas with concurrent demodicosis and dermatophytosis in a
cat. Aust Vet. 2001;79(7):470–5.
77. Zerbe CA, Nachreiner RF, Dunstan RW, et al. Hyperadrenocorticism in a cat. J Am Vet Med
Assoc. 1987;190(5):559–63.
78. Chalmers S, Schick RO, Jeffers J. Demodicosis in two cats seropositive for feline immunodeficiency virus. J Am Vet Med Assoc. 1989;194(2):256–7.
79. Guaguère E, Olivry T, Delverdier-Poujade A, et al. Demodex cati infestation in association
with feline cutaneous squamous cell carcinoma in situ: a report of five cases. Vet Dermatol.
1999;10(1):61–7.
80. Bailey RG, Thompson RC, Nickels DG. Demodectic mange in a cat. Aust Vet J.
1981;57(1):49.
81. Beale K. Feline demodicosis: a consideration in the itchy or overgrooming cat. J Feline Med
Surg. 2012;14(3):209–13.
82. Silbermayr K, Joachim A, Litschauer B, et al. The first case of Demodex gatoi in Austria,
detected with fecal flotation. Parasitol Res. 2013;112(8):2805–10.
83. Mueller RS. Treatment protocols for demodicosis: an evidence-based review. Vet Dermatol.
2004;15(2):75–89.
84. Johnstone IP. Doramectin as a treatment for canine and feline demodicosis. Aust Vet Pract.
2002;32(3):98–103.
85. Short J, Gram D. Successful treatment of Demodex gatoi with 10% Imidacloprid/1%
Moxidectin. J Am Anim Hosp Assoc. 2016;52(1):68–72.
86. Matricoti I, Maina E. The use of oral fluralaner for the treatment of feline generalised demodicosis: a case report. J Small Anim Pract. 2017;58(8):476–9.
87. Duangkaew L, Hoffman H. Efficacy of oral fluralaner for the treatment of Demodex gatoi in
two shelter cats. Vet Dermatol. 2018;29(3):262.
88. Wall R, Shearer D. Lice. In: Veterinary Ectoparasites: biology, pathology & control. 2nd ed.
Oxford: Blackwell Science; 2001. p. 162–78.
89. Pollmeier M, Pengo G, Longo M, et al. Effective treatment and control of biting lice, Felicola
subrostratus (Nitzsch in Burmeister, 1838), on cats using fipronil formulations. Vet Parasitol.
2004;121(1–2):157–65.
90. Shanks DJ, Gautier P, McTier TL, et al. Efficacy of selamectin against biting lice on dogs and
cats. Vet Rec. 2003;152(8):234–7.
91. Ketzis JK, Dundas J, Shell LG. Lynxacarus radovskyi mites in feral cats: a study of diagnostic methods, preferential body locations, co-infestations and prevalence. Vet Dermatol.
2016;27:425–e108.
92. Clare F, Mello RMLC. Use of fipronil for treatment of Lynxacarus radovskyi in outdoor cats
in Rio de Janeiro (Brazil). Vet Dermatol. 2004;15(Suppl 1):50. (abstract)
VetBooks.ir
436
F. Leone and H. S. Han
93. Han HS, Noli C, Cena T. Efficacy and duration of action of oral fluralaner and spot-on moxidectin/imidacloprid in cats infested with Lynxacarus radovskyi. Vet Dermatol. 2016;27:474–e127.
94. Catts EP, Mullen G. Myiasis (Muscoidea, Oestroidea). In: Mullen G, Durden L, editors.
Medical and veterinary entomology. Orlando: Academic Press; 2002. p. 317–43.
95. Fang Fang, Qinghua Chang, Zhaoan Sheng, Yu Zhang, Zhijuan Yin, Jacques Guillot,
Chrysomya bezziana: a case report in a dog from Southern China and review of the Chinese
literature. Parasitology Research.
96. Hock Siew Han, Peik Yean Toh, Hock Binn Yoong, Hooi Meng Loh, Lee Lee Tan, Yin Yin Ng,
(2018) Canine and feline cutaneous screw-worm myiasis in Malaysia: clinical aspects in 76
cases. Vet Dermatol. 29(5):442–e148.
97. Clarissa P de Souza, Guilherme G. Verocai, Regina HR Ramadinha, (2010) Myiasis caused
by the New World screwworm fly (Diptera: Calliphoridae) in cats from Brazil: report of five
cases. J Feline Med Surg. 12(2):166–8.
98. Thaís R. Correia, Fabio B. Scott, Guilherme G. Verocai, Clarissa P. Souza, Julio I. Fernandes,
Raquel M.P.S. Melo, Vanessa P.C. Vieira, Francisco A. Ribeiro, (2010) Larvicidal efficacy
of nitenpyram on the treatment of myiasis caused by Cochliomyia hominivorax (Diptera:
Calliphoridae) in dogs. Vet Parasitol. 173(1–2):169–72.
99. Hock Siew Han, Charles Chen, Carlo Schievano, Chiara Noli, (2018) The comparative efficacy
of afoxolaner, spinosad, milbemycin, spinosad plus milbemycin, and nitenpyram for the treatment of canine cutaneous myiasis. Vet Dermatol. 29(4):312–e109.
VetBooks.ir
Flea Biology, Allergy and Control
Chiara Noli
Abstract
Fleas are the most common ectoparasites and flea-bite allergy can develop in
cats. The clinical signs are represented by pruritus, excoriations, self-induced
alopecia, manifestations of the eosinophilic granuloma complex and miliary dermatitis, which often, but not exclusively, involves the caudal dorsal and ventral
part of the body. The diagnosis is obtained with the clinical presentation and
response to flea control. Flea control is based on adulticides, which kill adult
fleas on the cat, and insect growth regulators (IGR), which inhibit the development of pre-adult stages in the environment.
Introduction
The flea species most frequently identified in cats is Ctenocephalides felis felis
(Fig. 1). A comprehensive review on its biology and ecology has recently been published [1]. Fleas can be a cause and/or vector of a variety of diseases such as anemia in heavily infested kittens, tapeworm infestations, Lyme disease, pest, viruses,
hemoparasites, cat-scratch disease and flea allergy [1, 2]. Recognition of some of
these conditions, such as tapeworm in the cat or cat-scratch disease in the owner, is
a sign of flea infestation, even if asymptomatic in the feline carrier host.
Flea-bite allergy is by far the most frequent disease caused by fleas in cats and its
prevalence depends on the geographical region and local parasite prevention habits.
In a recent multicenter European study, flea-bite allergy was found to account for
about one third of all feline pruritic cases [3].
C. Noli (*)
Servizi Dermatologici Veterinari, Peveragno, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_20
437
VetBooks.ir
438
C. Noli
Fig. 1 Microscopic aspect of
the cat flea Ctenocephalides
felis felis (4×)
Pathogenesis of Flea Allergy
Cats are bitten by fleas several times a day [4]. Fleas insert their mouthparts
through the epidermis in the dermis and suck blood from the capillaries. During
this procedure, they deposit up to 15 salivary proteins within the epidermis
and superficial dermis that soften tissues and prevent blood coagulation [5, 6].
Hypersensitivity to these proteins induces local edema and a cellular infiltrate,
which constitutes the erythematous papule that may follow the bite. There are no
specific studies yet that identify the precise allergenic components of flea saliva
relevant for naturally sensitized cats. One study suggests that FSA1 (feline salivary antigen-1) may be a major flea saliva antigen in experimentally sensitized
laboratory cats [7]. It is thought that non-allergic animals suffer little or no discomfort while being bitten and that only flea allergic subjects develop pruritus
and skin disease.
Little is known about the pathogenesis of flea allergy in cats. Most flea allergic
cats have immediate positive intradermal skin test reactions to flea allergens and
delayed, type 4 reactions have been also described [8, 9]. As in dogs, allergen-­
specific IgE can be found in the serum of flea allergic cats by means of ELISA [8,
10]. Late-phase IgE-mediated cellular response and cutaneous basophil hypersensitivity have not yet been identified in cats.
Results of a study on early sensitization of 12-week-old kittens, which developed only mild clinical signs (10/18 kittens), suggest that cats exposed to fleas
early in their life are less likely to develop flea allergy than cats exposed at a later
age [11]. The authors suggested that early ingestion of fleas could induce tolerance, as cats experimentally exposed to fleas orally tended to have minimal clinical
VetBooks.ir
Flea Biology, Allergy and Control
439
signs and lower in vivo and in vitro test scores, although this was not statistically
different from the controls [11]. In the same study, cats continuously exposed to
fleas from 16 to 43 weeks of age developed either immediate or late reactivity to
live flea challenge. However, the same cats were not all positive on intradermal or
serology testing. Immediate test reactivity was reported to persist for more than
90 days after experimental sensitization [7]. In a study specifically designed to
clarify the role of intermittent exposure to flea bites, it was concluded that it had
neither a protective nor a predisposing effect on the development of clinical signs
of flea allergy [12].
Clinical Appearance
There is no age, breed, or sex predilection for the development of flea-bite hypersensitivity. In most cases flea control is either completely lacking or incomplete or
wrongly performed. Clinical signs are usually worse in the warmer months, particularly at the end of the summer, when the flea population is at its highest point. In
addition, many owners stop administering flea control in the same period, as they
feel it is no longer needed.
Clinical signs of feline flea allergy are not different from those caused by
other allergies in cats and include, alone (75% of cats) or in combination, pruritus, miliary dermatitis, self-induced alopecia, eosinophilic plaque and eosinophilic granuloma, lip ulcer and head and neck excoriations [3]. Please refer to
Chapter, Feline Atopic Syndrome: Epidemiology and Clinical Presentation for
a more extensive description of these clinical presentations. All of these signs
could be reproduced in experimental sensitization studies [12]. Prevalence of
lesions in flea-bite allergy is detailed in Table 1 [3]. A multicentric study on 502
pruritic cats reported a preferred localization of pruritus and lesions of miliary
dermatitis on the caudal dorsum (Fig. 2) in cats with flea-bite hypersensitivity, if
compared with other allergies [3]. In the same study, non-dermatological signs,
such as conjunctivitis, rhinitis, vomiting, diarrhea, and soft feces, were observed
in 30% of cats with flea-­bite allergy, and otitis was observed in 3% [3].
Table 1 Prevalence of clinical sign of allergy in cats with flea bite hypersensitivity (reference
Hobi)
Clinical sign
Miliary dermatitis
Prevalence
35%
Symmetrical alopecia
39%
Head and neck pruritus and excoriations
Eosinophilic granuloma complex (including
eosinophilic granuloma, eosinophilic plaque and
lip ulceration)
38%
14%
Most frequent distribution
Caudal dorsum, caudal
thighs or generalized
Caudal dorsum and flank
Abdomen
Head and neck
Granuloma: mouth, chin,
caudal aspect of the hind
legs
Plaque: abdomen, groin
Lip ulcer: upper lips
VetBooks.ir
440
C. Noli
Fig. 2 Self-inflicted
lesions on the back of a cat
with flea-bite allergy
Fig. 3 Fleas and flea feces
found in the coat of a
non-allergic cat
Differential Diagnoses and Diagnostic Approach
A dermatological examination of a cat should always include search for fleas and
their feces, by means of a thorough fine-tooth combing of the whole patient (Figs. 3,
4 and 5). Flea feces are made of dry blood and can be easily recognized as they
will leave a brown halo on a white moistened paper towel. Fleas or flea feces are
not always found as cats are excellent groomers and can eliminate all fleas in a few
hours [13]. Furthermore, the number of eggs that fall off flea allergic cats in the
environment is lower, leading to a less obvious animal and environmental infestation [13]. For this reason, a lack of fleas or flea dirt in the coat does not exclude a
diagnosis of flea allergy. The main differential diagnoses of flea allergy are other
allergies, such as adverse reactions to food and environmental allergic dermatitis,
VetBooks.ir
Flea Biology, Allergy and Control
441
Fig. 4 Abundant flea feces
and some adult fleas
obtained by flea combing
in a flea-infested cat
Fig. 5 Microscopic aspect
of the same material of
Fig. 4: flea feces appear as
red, curled structures. They
are made by over 90% of
the cat’s dry blood. This is
an important parental
investment by the female
flea, as flea feces represent
the main nourishment for
the flea larvae
because they share all the abovementioned clinical manifestations. Other less frequent differentials are other parasitic diseases (Chapter, Ectoparasitic Diseases),
psychogenic alopecia (Chapter, Psychogenic Diseases), dermatophytosis (for miliary dermatitis) and rare, pruritic, immune-mediated, autoimmune and neoplastic
diseases.
A suspect diagnosis of flea allergy may be confirmed by performing an intradermal skin test. The flea allergen is injected (0.05 ml) intradermally together
with a negative (saline) and a positive (histamine) control and reactions are read
at 15 minutes and 48 hours. Current or recent administration of glucocorticoids or
antihistamines (2 weeks for short-acting glucocorticoids and antihistamines, up to
8 weeks for depot glucocorticoids) may cause false-negative results. False-positive
VetBooks.ir
442
C. Noli
reactions in normal cats have been described: in one study, 36% of clinically ­normal
cats that had been exposed to fleas had a positive immediate skin test reaction to
flea antigens [14].
A positive predictive value of 85–100% was reported in earlier studies [9, 12,
15], while a more recent study performed with three different extracts obtained
a sensitivity of 33% and a specificity of 78–100% [8]. In a study on experimental induction of flea hypersensitivity, the presence of positive immediate intradermal test reactions did not correlate with the development of clinical signs [11].
Allergens used in older studies were whole body flea extracts (1:1000 w/v), while
more recently flea saliva or purified salivary antigens have been developed for a
more sensitive in vivo test [5]. However, in experimentally induced feline flea-bite
allergy, results of intradermal testing with purified allergens were not superior to
crude extracts in the correlation with clinical signs [11, 12]. Furthermore, it is not
known if the concentration used (1/1000 w/v) extrapolated from dogs is optimal for
cats or if higher concentrations should be used [16].
In vitro serologic tests (ELISA) with whole body flea extracts or purified flea
saliva or recombinant flea saliva antigens are available for determination of allergen-­
specific IgE in the feline serum. The readers should be warned that these tests may
only identify animals with IgE-mediated disease and fail to diagnose those with a
delayed reaction only. Furthermore, there are normal cats which may have allergen-­
specific IgE in the absence of clinical disease [8, 11, 12]. Sensitivity and specificity
of serological tests performed with flea extracts were reported to be 88% and 77%,
respectively, in one study [8] and 77% and 72% in another study [15], with a low
positive predictive value of 0.58 in the latter one.
Flea saliva represents only 0.5% of whole flea extracts and in vitro tests performed in dogs with flea salivary antigens gave much better results than those
­performed with whole flea extracts [17]. In vitro test with salivary antigens and the
use of high-affinity receptors FcεR1α gave an overall accuracy of 82% and may
represent a more reliable tool for the diagnosis of flea allergy in cats [Mc Call].
In practice, the best approach to the correct diagnosis is to implement an effective ectoparasite control together with/followed by a good hypoallergenic diet. If
an improvement is obtained, a dietary challenge can differentiate between flea-bite
allergy and food hypersensitivity. If no improvement is obtained, then environmental allergy or other less frequent pruritic conditions can be taken into consideration
(Chapter, Feline Atopic Syndrome: Diagnosis for a detailed description of the diagnostic approach to the pruritic cat).
Treatment
Flea control is pivotal for an effective treatment of flea-bite allergy. Adult fleas are
obligate ectoparasites [4] and topical or systemic flea control on the cat is mandatory. However, the development of the life stages from egg through to pupa occurs
in the immediate domestic environment of the infested pet rather than on the host
and this requires adjunct environmental treatment [1, 4, 18]. Contact with other
VetBooks.ir
Flea Biology, Allergy and Control
443
cats is another source of infection. Unfortunately, in a survey conducted on owners of flea-infested animals, only 71% of dogs and 50% of cats had been treated
against fleas in the previous 12 months [19]. One of the most frequent challenges
of treating fleas is that many owners, particularly in case of absence of parasites
and feces on the cat’s coat, will be skeptical and feel offended if faced with the
assumption that there could be fleas on their pets and in their homes and will thus
be unwilling to perform a thorough flea control. Explaining that there is no need
to host high amounts of fleas to develop allergy and that only a well-conducted
all-year-round flea control is able to prevent flea infestation can increase owner
compliance.
The Flea Cycle and Ecology
Veterinarians should take time to thoroughly explain why and how to perform correct flea control. This begins with telling the owner something about the flea cycle
[1, 4]. Flea eggs are produced on the host and fall off within 8 hours of production. High egg counts have been found in places where the animal sleeps, eats, or
spends most of its time. The eggs hatch after 1–10 days (Fig. 6). The larvae live
freely in the environment and move actively under furniture and rugs, deep in carpet
fibers, or under organic debris (grass, branches, leaves), in order to avoid light. After
5–11 days, the larvae produce a silk-like cocoon for their protection and camouflage. Inside the cocoon the larvae develop into pupae and then in 5–9 days become
young adults. The fleas in the cocoon are very well protected from insecticides and
unfavorable environmental conditions and may survive in a quiescent state for up
to 50 weeks. If a potential host is there, the fleas exit the cocoon and rapidly jump
on it. If no host is available, the newly emerged fleas can survive several days (up to
a
b
Fig. 6 Flea feces, eggs, and larvae obtained from the environment (couch) of a flea-infested cat.
(4×)
VetBooks.ir
444
C. Noli
2 weeks) in the environment. If the fleas do not find a domestic animal, they often
bite humans before finding their preferred host. Adult fleas are permanent parasites
of animals. As soon as they land on a host, they begin to feed. The first eggs are
produced on the host after 36–48 hours. One single female is capable of producing
up to 40–50 eggs a day and up to 2000 eggs in about 100 days of life. The minimum
length of the whole cycle is 12–14 days, with an average of about 3–4 weeks in most
household conditions, in winter as well. Adult fleas account for only 1–5% of all
fleas in the cat’s environment, 95–99% of the fleas being the egg, larval, or pupal
stage. In fact, it is thought that in temperate climates, the house is the major source
of re-infestation of small animals.
Flea Control
Factors important for successful flea control are efficacy and safety of the active
ingredient, possibly with long residual activity. Molecules effective on fleas usually belong to one of two categories: either they kill adult fleas (adulticides) or
they inhibit pre-adult stage development (insect growth regulators). Adulticides are
needed on the animal, in order to kill adult fleas on the host, ideally before they
bite and elicit the allergic reaction. Adulticides alone kill only 1–5% of the flea
population and do not stop environmental (household) infestation, i.e., eggs, larvae
and pupae, representing 95–99% of the entire flea population. Insect growth regulators are able to inhibit development of eggs and larvae and decrease environmental
infestation, but cannot prevent the allergic animal from being bitten by an adult flea
which comes from “outside” the house. Therefore both product types are needed
together for effective flea control, especially for flea allergic animals, in order to
break the flea life cycle in at least two stages. A list of antiparasitic products available on the market for cats, with their characteristics, is provided in Table 2.
Published trials on flea control measures have been recently extensively reviewed
by Rust [1]. The best way to quickly and surely eliminate infestation in a cat is by
administering an oral parasiticide. Nitenpyram is the most rapid one, as its effect
is seen as soon as 15–30 minutes after administration [20]. Nitenpyram is thus an
excellent means of diagnosing the presence of fleas, if given as soon as the cat enters
the clinic, as fleas can be seen falling on the table during the consultation. However,
being its duration of effect so short (48 hours in cats), it is not very practical as a
flea prevention means (the drug should be administered every 48–72 hours). Other
oral flea control products, with a slower onset of efficacy (8–12 hours) but with the
advantage of a monthly duration, are spinosad and lotilaner [21, 22]. In one study,
oral products were considered more effective than topical spot-ons applied by the
owner in dogs [23], probably due to better reliability of the mode of administration.
There is no data on cats.
Other common flea control measures are spot-on formulations containing an adulticide (imidacloprid, fipronil, selamectin, metaflumizone, dinotefuran, indoxacarb)
Flea Biology, Allergy and Control
445
VetBooks.ir
Table 2 Antiparasitic products available on the market for cats against fleas, at the time of writing
Name original Active
ingredient
producta
Frontline
Fipronil
Formulationb
Spot-on
Minimum
age of use
8 weeks
Fipronil
Methoprene
Fipronil
Pyriproxyfen
Fipronil
Methoprene
Eprinomectin
Praziquantel
Spot-on
8 weeks
Spot-on
10 weeks
Spot-on
7 weeks
Advantage
Advocate
Imidacloprid
Imidacloprid
Moxidectin
Spot-on
Spot-on
8 weeks
9 weeks
Stronghold/
Revolution
Selamectin
Spot-on
6 weeks
Stronghold
plus
Selamectin
Sarolaner
Spot-on
8 weeks
Comfortis
Spinosad
14 weeks
Activyl
Vectra felis
8 weeks
7 weeks
Fleas
Fleas
No
Yes
Bravecto
Indoxacarb
Dinotefuran
Pyriproxyfen
Fluralaner
Tablets (with
food!)
Spot-on
Spot-on
Parasitesc
Fleas, ticks, lice,
Cheyletiella
Fleas, ticks, lice,
Cheyletiella
Fleas, ticks, lice,
Cheyletiella
Fleas, lice, Otodectes,
Demodex, heartworm,
Notoedres, Cheyletiella,
Angiostrongylus, GE
nematodes, tapeworm
Fleas
Fleas, lice, Otodectes,
Demodex, heartworm,
Notoedres, Cheyletiella,
GE nematodes
Fleas, lice, Otodectes,
Demodex, heartworm,
Notoedres, Cheyletiella,
GE nematodes
Fleas, ticks, lice,
Otodectes, Demodex,
Cheyletiella, Notoedres,
heartworm, myiasis, GE
nematodes
Fleas, myiasis
Spot-on
(12 weeks)
8 weeks
No
Bravecto
plus
Fluralaner
Moxidectin
Spot-on
(12 weeks)
9 weeks
Credelio
Lotilaner
Tablet (with
food!)
8 weeks
Seresto/
Foresto
Imidacloprid
Flumethrin
Collar
(6–8 months)
10 weeks
Fleas, lice, ticks,
Otodectes, Demodex,
Notoedres, Cheyletiella,
myiasis
Fleas, ticks, lice,
Otodectes, Demodex,
Cheyletiella, Notoedres,
heartworm (8 weeks),
GE nematodes, myiasis
Fleas, ticks, lice,
Otodectes, Demodex,
Cheyletiella, Notoedres,
myiasis
Fleas, ticks, sandflies,
mosquitoes
Frontline
combo
Effipro Duo
Broadline
IGR
effect
No
Yes
Yes
Yes
Yes
Yes
Yes
Yes
No
No
No
Yes
(continued)
446
C. Noli
VetBooks.ir
Table 2 (continued)
Name original Active
ingredient
producta
Capstar
Nitenpyram
Formulationb
Tablet
(activity 72 h)
Minimum
age of use
4 weeks
Parasitesc
Fleas, myiasis
IGR
effect
No
The original/first product marketed with this ingredient is reported in the table. Depending on the
country, several other products are currently available containing fipronil, fipronil/methoprene,
fipronil/pyriproxyfen, and imidacloprid
b
Monthly administration unless stated otherwise
c
Both label and “off-label” parasites are reported in this table
a
to be administered between the shoulder blades every 4 weeks. Pulicidal efficacy
of each one of these drugs has been proven to be excellent (at least 90%) for up to
4 weeks in laboratory clinical trials [1]. Among these, indoxacarb is a pro-insecticide that must be bio-activated by insect enzymes to generate the active metabolite
able to kill fleas and ticks. In mammals, indoxacarb is metabolized to inactive molecules by the liver and is not toxic, so that it is designated by the US Environmental
Protection Agency as a “reduced risk” pesticide.
Recently, a new spot-on formulation for cats based on fluralaner, a member of a
new class of antiparasitic agents, the isoxazolines, has been marketed with a residual
activity against fleas for up to 3 months [24]. Fluralaner is absorbed transdermally
and redistributed systemically, so that fleas will need to bite the cat to be killed. A
3-month duration time probably improves owner’s compliance and can be preferred
in allergic subjects.
There is one flea collar registered for use in the cat containing 10% imidacloprid
and 4.5% flumethrin, with a 6–8-month-long pulicidal activity. This product has the
advantages of being less expensive than spot-ons or tablets, with a higher compliance and repellent efficacy against fleas, ticks, mosquitos and sandflies vectors of
leishmaniosis [25].
Some of the abovementioned insecticides also offer ovicidal and larvicidal
­activity (e.g., imidacloprid [26] or selamectin [27]), while others are formulated in
association with an IGR, such as pyriproxyfen or methoprene. Insect growth regulators (IGR) interfere with the development of pre-adult flea stages, which account
for the vast majority of the total flea population (up to 99%). They have a very
low toxicity for mammals, because they act on very insect-specific metabolic pathways. The idea behind administering a product with IGR effect on the animal is
that treated hairs shed in the environment are able to inhibit eggs’ hatching and/or
larval moulting. The use of an IGR is fundamental to reduce the environmental flea
population, thus the flea burden on the cat and the consequent clinical symptomatology. IGR sprays, containing methoprene or pyriproxyfen, can also be used in
the e­ nvironment, especially in case of heavy or recurrent infestation. The principal
strategic problem in trying to control a domestic flea population, however, is dealing
with young adult fleas within the protective pupal case [23]. These can yield live,
viable adults for periods of several months after all eggs, larvae and other adults
have been killed, and repeated applications of environmental treatment may be necessary in some cases. Recently a 0.4% environmental dimeticone spray was able to
VetBooks.ir
Flea Biology, Allergy and Control
447
prevent emergence of young adult fleas from cocoons and proved to be efficacious
in immobilizing larvae and adults in the environment [28], with efficacy persisting
for more than 3 weeks.
Certain physical measures can assist in flea control. Washable surfaces can be
cleaned to remove organic matter and flea feces on which larvae feed. Vacuum
cleaning will remove 20% of larvae and up to 60% of eggs as well as flea feces and
organic matter. Vacuum cleaning assists spray penetration by raising the fibers in
carpets. Bedding and other washable items should be laundered at the highest temperature possible. Carpets and soft furnishings should not be washed as increased
humidity favors larval development.
How to Perform an Effective Flea Control and Causes of Failure
An adulticide has to be applied to every animal in the household all year round and
an IGR has either to be applied in the environment or to all pets. Flea control must be
thoroughly and constantly applied in order to be effective; thus, client compliance is
the most important element for a successful flea control. Recurrence of signs usually
depend on lack in flea control, which might be due to one or more of these factors [29]:
––
––
––
––
Use of ineffective products
Insufficient dosage or lack of application in the whole house or on all animals
Use of adulticides without IGR or IGR without adulticides
Too long period of time between administrations
Questioning the owner about how they perform flea control will nearly always
identify the problem and it is our task to explain and convince them about the importance of a complete flea control.
Although flea control is mandatory, it may not be sufficient to result in complete
control of the dermatosis in all cases, particularly where there is continued contact
with untreated individuals. In such cases, anti-pruritic treatment will be necessary.
Please refer to Chapter, Feline Atopic Syndrome: Therapy for a detailed discussion
of anti-pruritic drugs in cats.
The potential for vaccination, either against the immunogenic salivary proteins
of the flea or against concealed antigens within the flea gut, has been explored
with variable results and could offer possibilities for the future management of flea
allergy [7, 30–32].
Conclusion
Flea-bite hypersensitivity is one of the most important allergic skin conditions in
cats, which can manifest with different clinical signs and has many possible differential diagnoses. Intradermal and in vitro allergy tests are not always reliable
diagnostic tools and rigorous flea control, by means of adulticides and insect growth
regulators, represents the best tool for diagnosing and treating this condition.
448
C. Noli
VetBooks.ir
References
1. Rust MK. The biology and ecology of cat fleas and advancements in their Pest management: a
review. Insects. 2017;8:118.
2. Shaw SE, Birtles RJ, Day MJ. Arthropod transmitted infectious diseases of cats. J Feline Med
Surg. 2001;3:193–209.
3. Hobi S, Linek M, Marignac G, et al. Clinical characteristics and causes of pruritus in
cats: a multicentre study on feline hypersensitivity-associated dermatoses. Vet Dermatol.
2011;22:406–13.
4. Dryden MW, Rust MK. The cat flea: biology, ecology and control. Vet Parasitol. 1994;52:1–19.
5. Frank GR, Hunter SW, Stiegler GL, et al. Salivary allergens of Ctenocephalides felis: collection, purification and evaluation by intradermal skin testing in dogs. In: Kwochka KW,
Willemse T, von Tscharner C, editors. Advances in veterinary dermatology, volume 3. Oxford:
Butterworth Heinemann; 1998. p. 201–12.
6. Lee SE, Johnstone IP, Lee RP, et al. Putative salivary allergens of the cat flea, Ctenocephalides
felis felis. Vet Immunol Immunopathol. 1999;69:229–37.
7. Jin J, Ding Z, Meng F, et al. An immunotherapeutic treatment against flea allergy dermatitis in
cats by co-immunization of DNA and protein vaccines. Vaccine. 2010;28:1997–2004.
8. Bond R, Hutchinson MJ, Loeffler A. Serological, intradermal and live flea challenge tests in
the assessment of hypersensitivity to flea antigens in cats (Felis domesticus). Parasitol Res.
2006;99:392–7.
9. Lewis DT, Ginn PE, Kunkle GA. Clinical and histological evaluation of immediate and
delayed flea antigen intradermal skin test and flea bite sites in normal and flea allergic cats. Vet
Dermatol. 1999;10:29–38.
10. McCall CA, Stedman KE, Bevier DE, Kunkle GA, Foil CS, Foil LD. Correlation of feline IgE,
determined by Fcε RIα-based ELISA technology, and IDST to Ctenocephalides felis salivary
antigens in a feline model of flea bite allergic dermatitis. Compend Contin Educ Pract Vet.
1997;19(Suppl. 1):29–32.
11. Kunkle GA, McCall CA, Stedman KE, Pilny A, Nicklin C, Logas DB. Pilot study to assess the
effects of early flea exposure on the development of flea hypersensitivity in cats. J Feline Med
Surg. 2003;5:287–94.
12. Colombini S, Hodgin EC, Foil CS, Hosgood G, Foil LD. Induction of feline flea allergy
dermatitis and the incidence and histopathological characteristics of concurrent indolent lip
ulcers. Vet Dermatol. 2001;12:155–61.
13. McDonald BJ, Foil CS, Foil LD. An investigation on the influence of feline flea allergy on the
fecundity of the cat flea. Vet Dermatol. 1998;9:75–9.
14. Moriello KA, McMurdy MA. The prevalence of positive intradermal skin test reactions to lea
extracts in clinically normal cats. Comp Anim Pract. 1989;19:28–30.
15. Foster AP, O’Dair H. Allergy skin testing for skin disease in the cat in vivo vs in vitro tests. Vet
Dermatol. 1993;4:111–5.
16. Austel M, Hensel P, Jackson D, et al. Evaluation of three different histamine concentrations in
intradermal testing of normal cats and attempted determination of the irritant threshold concentrations of 48 allergens. Vet Dermatol. 2006;17:189–94.
17. Cook CA, Stedman KE, Frank GR, Wassom DL. The in vitro diagnosis of flea bite hypersensitivity: flea saliva vs. whole flea extracts. In: Proceedings of the 3rd veterinary dermatology
world congress, 1996 Spet 11–14. Edinburgh; 1996. p. 170.
18. Osbrink WLA, Rust MK, Reierson DA. Distribution and control of cat fleas in homes in
Southern California (Siphonaptera: Pulicidae). J Med Entomol. 1986;79:135–40.
19. Peribáñez MÁ, Calvete C, Gracia MJ. Preferences of pet owners in regard to the use of insecticides for flea control. J Med Entomol. 2018;55:1254–63.
20. Dobson P, Tinembart O, Fisch RD, Junquera P. Efficacy of nitenpyram as a systemic flea adulticide in dogs and cats. Vet Rec. 2000;147:709–13.
VetBooks.ir
Flea Biology, Allergy and Control
449
21. Cavalleri D, Murphy M, Seewald W, Nanchen S. A randomized, controlled field study to
assess the efficacy and safety of lotilaner (Credelio™) in controlling fleas in client-owned cats
in Europe. Parasit Vectors. 2018;11:410.
22. Paarlberg TE, Wiseman S, Trout CM, et al. Safety and efficacy of spinosad chewable tablets
for treatment of flea infestations of cats. J Am Vet Med Assoc. 2013;242:1092–8.
23. Dryden MW, Ryan WG, Bell M, et al. Assessment of owner-administered monthly treatments
with oral spinosad or topical spot-on fipronil/(S)-methoprene in controlling fleas and associated pruritus in dogs. Vet Parasitol. 2013;191:340–6.
24. Bosco A, Leone F, Vascone R, et al. Efficacy of fluralaner spot-on solution for the treatment of
ctenocephalides felis and otodectes cynotis mixed infestation in naturally infested cats. BMC
Vet Res. 2019;15:28.
25. Brianti E, Falsone L, Napoli E, et al. Prevention of feline leishmaniosis with an imidacloprid
10%/flumethrin 4.5% polymer matrix collar. Parasit Vectors. 2017;10:334.
26. Jacobs DE, Hutchinson MJ, Stanneck D, Mencke N. Accumulation and persistence of flea
larvicidal activity in the immediate environment of cats treated with imidacloprid. Med Vet
Entomol. 2001;15:342–5.
27. McTier TL, Shanks DJ, Jernigan AD, Rowan TG, Jones RL, Murphy MG, et al. Evaluation
of the effects of selamectin against adult and immature stages of fleas (Ctenocephalides felis
felis) on dogs and cats. Vet Parasitol. 2000;91:201–12.
28. Jones IM, Brunton ER, Burgess IF. 0.4% dimeticone spray, a novel physically acting household treatment for control of cat fleas. Vet Parasitol. 2014;199:99–106.
29. Halos L, Beugnet F, Cardoso L, et al. Flea control failure? Myths and realities. Trends Parasitol.
2014;30:228–33.
30. Heath AW, Arfsten A, Yamanaka M, et al. Vaccination against the cat flea Ctenocephalides felis
felis. Parasite Immunol. 1994;16:187–91.
31. Halliwell REW. Clinical and immunological response to alum-precipitated flea antigen in
immunotherapy of flea-allergic dogs: results of a double blind study. In: Ihrke PJ, Mason IS,
White SD, editors. Advances in veterinary dermatology, vol. 2. Oxford: Pergamon Press; 1993.
p. 41–50.
32. Kunkle GA, Milcarsky J. Double-blind flea hyposensitization trial in cats. J Am Vet Med
Assoc. 1985;186:677–80.
VetBooks.ir
Feline Atopic Syndrome: Epidemiology
and Clinical Presentation
Alison Diesel
Abstract
Although very well defined and characterized in the dog, feline atopic syndrome
remains less well understood with regard to disease pathogenesis and clinical
presentations. While many similarities exist, questions remain whether atopic
dermatitis is the same disease entity in dogs and cats. Atopic dermatitis in the cat
is often referred to as “feline atopic syndrome” or “non-flea, non-food hypersensitivity dermatitis (NFNFHD).” Although the diagnostic process is similar for
dogs and cats, with both being a diagnosis of exclusion, demonstration of immunoglobulin-­E (IgE) involvement in feline atopic syndrome has been inconclusive. As with canine atopic dermatitis, pruritus remains a feature of the disease in
cats; however, the distribution of pattern of pruritus and lesions is more variable
in feline patients. Cats with feline atopic syndrome will typically present with at
least one of four common cutaneous reaction patterns (head/neck/pinnal pruritus
with excoriations, self-induced alopecia, miliary dermatitis, eosinophilic skin
lesions). Additionally, non-cutaneous clinical signs may also be observed.
Introduction
Although very well defined and characterized in dogs and humans, feline atopic syndrome remains less well understood with regard to disease pathogenesis and clinical presentations. While many similarities exist, questions remain whether atopic
dermatitis is the same disease entity in dogs and cats. In general, when allergic
skin disease is compared across the two species, much less is ­known/documented
A. Diesel (*)
College of Veterinary Medicine and Biomedical Sciences, Texas A&M University,
College Station, TX, USA
e-mail: ADiesel@cvm.tamu.edu
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_21
451
VetBooks.ir
452
A. Diesel
in cats, especially in regard to atopic dermatitis. While the term “feline atopy” has
been a part of the veterinary literature since 1982 [1], this terminology has fallen
out of favor when discussing the disease in cats. “Feline atopic dermatitis” was used
initially to describe a clinical syndrome in feline patients with recurrent pruritic skin
disease, positive reactions to several environmental allergens on intradermal testing
and where other causes of pruritus (e.g., external parasites, infections) had been
ruled out. Due to the lack of conclusive demonstration of immunoglobulin-E (IgE)
involvement in the disease process, most veterinary dermatologists prefer either
“feline atopic syndrome” (FAS) or “non-flea, non-food hypersensitivity dermatitis”
(NFNFHD) when referring to what was historically referred to as feline atopic dermatitis (AD) [2].
While the condition remains a diagnosis of exclusion in both species, feline
atopic syndrome presents a unique set of challenges for the veterinary practitioner.
This includes not only quandaries in interpretation of diagnostic tests but also evaluation of the particular clinical syndromes unique to the feline patient and currently
limited options for therapeutic intervention compared to the canine counterpart.
This chapter aims to discuss what is presently known with regard to the pathogenesis of feline atopic syndrome, the epidemiology of disease, and observed clinical
presentations. Subsequent chapters will present a discussion on diagnostic evaluations and current therapy.
Pathogenesis of Feline Atopic Syndrome
Compared to dogs and people where the pathogenesis of atopic dermatitis is relatively well characterized [3–5], there remains a paucity of information present in the
literature with regard to the development of feline atopic syndrome. Although the
body of information continues to grow in certain areas of disease pathogenesis for
dogs and people (particularly in regard to influences in barrier function and more
specific immunological factors), many of these foci have not yet been explored for
the allergic feline patient. What has been documented, however, can be discussed
with regard to the historical classic triad of factors involved in the development of
atopic dermatitis (genetic influence, environmental factors, immunological abnormalities) and influences of barrier function on the course of disease.
Genetic Factors
In dogs and people, it is relatively well established that a genetic predisposition will
often contribute to an allergic phenotype, specifically in relation to the development of atopic dermatitis. This has been shown in several human twin studies [6]
and in the evaluation of the influence of filaggrin mutation as a contributory factor [7]. In the dog, specific phenotypes have been described for several commonly
affected canine breeds [8]; however as in people, it is clear genetics is only part of
the picture. The complex genotype of canine atopic dermatitis, with multiple genes
VetBooks.ir
Feline Atopic Syndrome: Epidemiology and Clinical Presentation
453
involved in the genetic component of the disease development, indeed speaks of
the multifaceted nature of the disease. That said, with certain documented genetic
variations and improved understanding of the genetic influence for certain patients,
targeted therapy aimed at specific molecules may be able to be developed and
implemented in the future [9].
In the cat, however, genetic influence in the development of feline atopic syndrome has been only loosely documented [10]. While it seems plausible that indeed
there is a genetic component to the disease in cats, to what degree this is apparent is
far from known at this time.
Environmental Factors
As is seen with atopic dermatitis in dogs and people, exposure to environmental
allergens exacerbates clinical signs in cats with atopic syndrome [11]. This is apparent in the naturally occurring disease presentation and has been supported with a
clinical model. In a study utilizing a modified patch test with aeroallergens applied
to the skin of healthy and allergic cats, only cats with atopic syndrome developed
an inflammatory infiltrate similar to that seen in the lesional skin of cats with the
spontaneous disease [12]. Whether application or exposure to aeroallergens in a
laboratory setting would lead to more generalized lesions associated with atopic
syndrome in the cat, as it does in dogs [13], has not been investigated.
Although a positive “allergy test” does not diagnose atopic dermatitis in any
known species, the historical definition of atopic dermatitis in the cat [1] included
the description of cats with several positive reactions to environmental allergens on
intradermal allergen tests. Intradermal allergen testing (as well as serum allergen
testing) for environmental allergens remains a cornerstone of support for the clinical diagnosis of feline atopic syndrome (see further discussion in Chapter, Feline
Atopic Syndrome: Diagnosis). This combined with a favorable response to allergen
immunotherapy in many cats with atopic syndrome further supports the influence of
environmental factors in disease pathogenesis.
Immunological Findings in Cats with Atopic Syndrome
Pulling together all aspects of the disease, the current definition of canine atopic
dermatitis describes “a genetically predisposed inflammatory and pruritic allergic
skin disease with characteristic clinical features associated with IgE antibodies most
commonly directed against environmental allergens” [14]. The influence of IgE has
clearly been demonstrated in this species as well as in people; however, this association is less well defined for feline atopic syndrome. Indeed, the role of IgE remains
an area of contention with regard to the immunological factors lending to disease
development. Part of the argument stems from a lack of correlation with serum IgE
levels in cats and clinical disease [15]; however, levels of allergen-specific IgE do
not always correlate with clinical disease in canine atopic dermatitis either [16].
VetBooks.ir
454
A. Diesel
There is a reasonable body of evidence, however, that supports the influence of IgE
in hypersensitivity dermatitis in the cat. Passive cutaneous anaphylaxis testing has
been used in cats to demonstrate the transfer of allergen-specific cutaneous reactivity from a sensitized/allergic cat to a naïve feline via injection of serum from the
allergic individual [17, 18]. This reactivity, however, does not occur if the serum is
heated prior to injection. The heating process inactivates IgE but not other antibodies, thereby supporting IgE involvement [17, 19, 20]. When anti-IgE is injected
into the skin of normal cats, immediate and delayed inflammatory responses occur
[21], sharing many macroscopic and microscopic features of what has previously
been reported in cats with spontaneously occurring allergic skin disease [10, 15]. A
similar inflammatory response, however, was not observed with injection of IgG in
this group [21], again supporting the involvement of IgE in feline hypersensitivity
dermatitis. The role of IgE has been well established in other allergic diseases in
the cat, most notably feline asthma [20, 22]. Given this condition occurs not infrequently in cats with (presumed) allergic skin disease [23], the suspected role of IgE
in the phenotype of both conditions cannot be ignored.
Although there is still a bit of uncertainty in regard to the immunopathogenesis
of feline atopic syndrome, there is a similar pattern of inflammatory infiltrate in
the skin of allergic cats compared to that which is seen in humans and dogs with
chronic atopic dermatitis [24]. Certain cell types involved in the innate and adaptive
immune system can be seen in altered numbers in the skin of allergic cats compared
to those without hypersensitivity dermatitis. Dendritic cells, including Langerhans
cells, have been reported in higher numbers in allergic feline skin [24, 25]. These
cells interface with the environment, lending to development of allergic inflammation, and have been implicated as well in the generation of atopic dermatitis in
people [26]. Eosinophils, often seen with various allergic diseases across multiple
species, are additionally increased in the skin of cats with allergy. Indeed, these
cells are a conspicuous infiltrate in inflammatory lesions of feline allergic dermatitis, particularly in miliary dermatitis lesions, and are suspected to be the more
specific indicator of a hypersensitivity response in cutaneous allergy in cats [27].
Tissue inflammation occurs secondary to the release of granule contents, including
major basic protein, as well as inflammatory cytokine expression [28]. Although
not specific to hypersensitivity dermatitis in the cat, mast cells are often increased
in the skin of cats with allergies compared to healthy cat skin [27]. Additionally, as
is seen in people with atopic dermatitis [29], mast cells in allergic cat skin undergo
a change in granule content. In cats with allergic skin disease, a markedly lower
number of mast cells have been observed staining for tryptase as opposed to chymase [27]. This is compared to healthy cat skin where all mast cells can be seen with
tryptase staining and approximately 90% observed when staining for chymase [30].
It has been well documented that a skewed T-cell response in favor of T helper
2 (Th2) over Th1 is a part of the immunological development of atopic dermatitis
in dogs and people. T cell involvement also appears to be involved in the immunopathogenesis of feline atopic syndrome. This has been seen with histopathological studies documenting increased populations of CD4+ T cells in allergic cat skin
compared to that of CD8+; these cells are generally not observed in the skin from
healthy cats [31]. Additionally, an increased number of IL-4 producing T cells have
VetBooks.ir
Feline Atopic Syndrome: Epidemiology and Clinical Presentation
455
been found in the skin of allergic cats compared to that of healthy controls, supportive of a Th2 infiltrate [32]. This skewed population of T cells has not, however,
been demonstrated in the peripheral blood of allergic cats compared to healthy controls [31]. The inflammatory cytokine profile has also not been well elucidated in
the skin or peripheral blood of cats with feline atopic syndrome. Differences in the
gene expression of various inflammatory interleukins and other cytokines could not
be detected when comparing the skin of normal, lesional, and non-lesional allergic
cat skin [33]. More recently, increased circulating levels of IL-31 have been demonstrated in sera from allergic cats compared to those without allergic skin disease
[34] as has been shown in canine atopic dermatitis. This suggests involvement of
this inflammatory cytokine in feline allergic dermatoses; however, a causative role
has yet to be determined.
Skin Barrier and Other Factors
The role of barrier function in the skin of people and dogs with atopic dermatitis
has become an increasingly important area of investigation. This factor, however,
has not been well explored in cats with feline atopic syndrome. One study observed
differences in transepidermal water loss (TEWL), skin hydration, and pH at various
body sites in healthy cats [35]. Recently, a study examined the relationship between
TEWL and severity of clinical symptoms in cats with feline atopic syndrome [36].
Using two scoring systems to assess skin lesions in allergic cats (Scoring Feline
Allergic Dermatitis (SCORFAD) and Feline Extent and Severity Index (FeDESI)),
a positive correlation was observed between TEWL and severity of clinical lesions
at certain body sites, particularly when using the SCORFAD measurements. Less
association was observed with FeDESI scoring. While there may indeed be differences in TEWL in allergic cats compared to healthy controls, the measurements may
be less useful compared to what is seen in dogs and humans with atopic dermatitis.
In people and dogs with atopic dermatitis, bacterial infection and yeast overgrowth can exacerbate clinical signs of disease. The same appears to be true in
some cats with feline atopic syndrome; secondary infections, however, with either
bacteria or yeast tend to occur less frequently in allergic cats compared to allergic
dogs or people. Although the exact implications have yet to be determined, there
is a growing body of evidence documenting changes in the microbiome in atopic
individuals. Indeed, this has been reported in both humans [37] and dogs [38], and
more recently in allergic cats compared to healthy controls [39]. While there are
some similarities across species (e.g., the Staphylococcus species is more abundant
in allergic individuals compared to healthy controls), there are additionally species differences. Contrary to allergic dogs and humans, allergic cats seem to retain
microbial diversity, in that the number of bacterial species was not significantly
different in allergic compared to healthy individuals [39]. Furthermore, compared
to dogs and people where differences in bacterial communities are seen at specific body locations in the face of an allergic “flare,” in allergic cats, their entire
body becomes colonized by an altered bacterial population independent of location
sampled. This is postulated to be due to the fastidious grooming behavior of cats.
VetBooks.ir
456
A. Diesel
These differences may partially explain why secondary infections are less common
in allergic cats compared to what is observed in dogs in people. What implication
this dysbiosis has in disease development and/or response to therapeutic intervention remains yet to be discovered.
Epidemiology of Feline Atopic Syndrome
The exact prevalence of feline atopic syndrome in the general population has not
been well described in the veterinary literature. A retrospective study on the population of cats seen at a teaching hospital in the United States identified “allergies”
accounting for 32.7% of the feline skin diseases presented to the hospital during a
15-year period. “Atopic dermatitis” itself represented 10.3% of the feline dermatoses
observed [40]. A similar study over a 1-year period at a university teaching hospital
in Canada diagnosed “atopic dermatitis” in 7 of 111 (6.3%) presented for evaluation
of dermatological disease [41]. In another study evaluating dermatological diseases
seen in general practice in the United Kingdom, however, only 2 out of 154 (1.3%)
cats were diagnosed with “atopic dermatitis.” It is important to note, however, that
other cutaneous reaction patterns (e.g., miliary dermatitis, eosinophilic granuloma
complex) were observed in this population without a defined etiology [42]. This
difference of prevalence may also partly be explained by differences in diagnoses
obtained by a general practitioner compared to a specialist in dermatology.
Clinical Presentation of Feline Atopic Syndrome
As with canine atopic dermatitis, clinical signs of feline atopic syndrome revolve
around the presence of pruritus in the cat. Comparatively, however, the distribution
of pruritus and lesions is less well defined in the feline patient. With dogs, clinical signs of canine atopic dermatitis typically follow a very predictable pattern to
include the face, concave pinnae, axillary and inguinal folds, ventrum, perineal skin,
flexural surfaces, and paws [43, 44]. With cats, however, pruritus and lesions will
generally include any one or more of the commonly recognized cutaneous reaction patterns reflecting a response to inflammation in feline skin [2]. While these
patterns do not reflect a specific etiology, they often are indicative of underlying
allergic skin disease.
Head/Neck/Pinnal Pruritus with Excoriations
Also referred to as cervicofacial pruritic dermatitis, lesions associated with this
reaction pattern are restricted to the front part of the cat. From the neck directed
caudally, the cat will generally appear normal. The face, ears, and neck, however,
may be marked with excoriation, crusts, alopecia, and erythema (Fig. 1). In some
cases, pruritus can be so severe that obvious self-trauma is apparent.
VetBooks.ir
Feline Atopic Syndrome: Epidemiology and Clinical Presentation
457
Fig. 1 Cat with
cervicofacial pruritus
secondary to feline atopic
syndrome
Fig. 2 Cat with selfinduced alopecia
secondary to feline atopic
syndrome. Note barbered
hair over site of forelimb
amputation aligning with
contralateral axillary
alopecia
Self-Induced Alopecia
Historically, cats with self-induced alopecia (often referred to as “symmetrical
alopecia,” “fur mowing,” or “barbering”) have been overdiagnosed with behavioral abnormalities and psychogenic alopecia. With this reaction patterns, cats will
remove hair by excessive licking, chewing, or pulling to the point of partial to near
complete alopecia of the affected body region (Fig. 2). The hairs will frequently
appear broken and rough where over-grooming has occurred. Concurrent erythematous skin and excoriation may or may not be present.
Miliary Dermatitis
Deriving its name from millet seeds (small grains), miliary dermatitis lesions in
the cat will often better be palpated as opposed to visualized. Lesions most commonly present along the neck and dorsum; however, the sparsely haired region of
458
b
VetBooks.ir
a
A. Diesel
Fig. 3 (a) Miliary dermatitis lesions on dorsum of cat with flea allergy dermatitis. (b) Miliary
dermatitis lesions on head of cat with feline atopic syndrome
preauricular skin can be the best location to visualize miliary dermatitis in the feline
patient without having to clip the hair coat (Fig. 3a, b). When present, the lesions
appear as small, pinpoint erythematous crusted papules. On palpation, the lesions
will feel like small grits or grains under the skin, as if petting coarse sandpaper.
Eosinophilic Skin Lesions
Included in this group of lesions are eosinophilic granulomas, eosinophilic plaques,
and lip (“indolent,” “rodent”) ulcers. This collection of lesions used to be referred
to as the feline “eosinophilic granuloma complex”; however, this terminology has
fallen out of favor with many dermatologists when describing these lesions in the
cat due to their distinct clinical and histopathological appearance.
While they can appear on any given body surface, eosinophilic granulomas may
appear most frequently on the caudal thigh (Fig. 4a) or the ventral surface of the
chin (Fig. 4b). The previous may be referred to as “linear granulomas,” while the
latter may be termed “fat chin” or “pouty” cat lesions. Granulomas are typically
semi-firm, rather well circumscribed, and may be seen in the presence or absence of
pruritus. Additionally, granulomas may occur in the oral cavity secondary to feline
atopic syndrome (Fig. 4c). Cats may initially present with clinical signs of dysphagia, drooling, decreased appetite, or even dyspnea depending on the size of the
lesion present. Alternatively, they may be found on oral examination in the absence
of any obvious clinical abnormalities.
Lip (indolent) ulcers may also present in the absence of clinical signs. These
craterous, ulcerative lesions may be unilaterally or bilaterally present on the upper
lips of affected cats (Fig. 5). Extension along the philtrum to the nasal planum is a
fairly common finding.
Of the three lesions, eosinophilic plaques tend to be associated with severe pruritus and concurrent self-induced alopecia. The lesions may again be present on
any part of the body and are most commonly visualized on the ventral abdomen.
Feline Atopic Syndrome: Epidemiology and Clinical Presentation
b
VetBooks.ir
a
459
c
Fig. 4 (a) Eosinophilic granuloma lesions on caudal thighs of a cat with feline atopic syndrome.
(b) Eosinophilic granuloma lesion on the chin of a cat with feline atopic syndrome. (c) Eosinophilic
granuloma lesion in caudal oral cavity of cat with feline atopic syndrome. This cat had moderate
dysphagia and respiratory stridor due to the size of the lesion
Fig. 5 Bilateral indolent
ulcer on upper lip of a cat
with feline atopic
syndrome
VetBooks.ir
460
A. Diesel
Eosinophilic plaques are generally well-circumscribed, erythematous plaque-like
lesions with a glistening, moist surface. Lesions are often multifocal and may
coalesce into a larger, single plaque (Fig. 6).
Extra-cutaneous Clinical Signs
While dermatological manifestation is the hallmark of feline atopic syndrome, other
extra-cutaneous clinical signs may also present in the allergic cat. This may include
allergic otitis, sinusitis, and conjunctivitis as well as feline small airway disease
(“feline asthma”) in certain patients. How frequently these diseases/clinical manifestations occur concurrently, however, is unknown.
As part of cervicofacial pruritic dermatitis, pinnal pruritus is a fairly common
clinical finding in cats with atopic syndrome. On otoscopic examination, however,
the external ear canals themselves are frequently normal in appearance. This is in
contrast to dogs with canine atopic dermatitis as they frequently present with erythematous otitis externa secondary to allergic disease [2]. Commonly mistaken for
ear mite infestation, many cats with atopic syndrome will present with recurrent
ceruminous otitis externa, often in the absence of infectious organisms such as bacteria or yeast. This can contribute to the otic pruritus observed.
Commonly reported in cats with feline atopic syndrome, sneezing may be indicative of sinusitis in allergic cats. Although uncertain the exact prevalence, some
sources site this concurrent clinical finding of upward of 50% in cats with feline
atopic syndrome [45]. While there is only a single report of feline allergic rhinitis
documented in the veterinary literature [46], this may be under-reported since a
Fig. 6 Large eosinophilic
plaque on the abdomen of
a cat with feline atopic
syndrome. Close
inspection of the lesion
shows where multiple
smaller plaques coalesced
to form the larger lesion
seen on the patient
VetBooks.ir
Feline Atopic Syndrome: Epidemiology and Clinical Presentation
461
clinical suspicion may be addressed in the absence of a definitive (e.g., biopsy)
diagnosis. However, the exact prevalence of these findings in cats with atopic syndrome is unknown since imaging studies have yet to investigate these concurrent
disease presentations.
As with other extra-cutaneous manifestations of allergies, the prevalence of
concurrent feline asthma in cats with atopic syndrome is uncertain. Feline small
airway disease or feline asthma is a complex syndrome; however, many cats have
an allergic pathogenesis [22]. In a pilot study evaluating the prevalence of positive
reactions to inhaled allergens in cats with small airway disease [23], the presence
of concurrent or pre-existing dermatologic abnormalities was quite high, making
recruitment of patients for the study difficult. This finding may indicate a higher
percentage of cats with both allergic airway disease and feline atopic syndrome. In
some cases, the severity of respiratory signs may, however, overshadow the presence of concurrent skin disease, or treatment for feline asthma (i.e., glucocorticoids) may control the signs of cutaneous allergy and thereby mask the true clinical
appearance. Further study is warranted to better elucidate the relationship between
the two disease conditions.
Conclusions
Feline atopic syndrome draws several parallels to canine atopic dermatitis mostly
in the involvement of pruritus in this clinical diagnosis. Much uncertainty remains,
however, in regard to the similarities which can be identified between the two allergic conditions in regard to clinical manifestations of disease and the particular
nature of disease pathogenesis. There remains quite a good amount of information yet to be discovered with regard to the patient with feline atopic dermatitis.
Compared to their canine counterpart, studies are lacking in the veterinary literature
for allergic cats.
References
1. Reedy LM. Results of allergy testing and hyposensitization in selected feline skin diseases. J
Am Anim Hosp Assoc. 1982;18:618–23.
2. Hobi S, Linek M, Marignac G, Olivry T, Beco L, Nett C, et al. Clinical characteristics and
causes of pruritus in cats: a multicentre study on feline hypersensitivity-associated dermatoses.
Vet Dermatol. 2011;22:406–13.
3. Marsella R, De Benedetto A. Atopic dermatitis in animals and people: an update and comparative review. Vet Sci. 2017;4(3):37.
4. Peng W, Novak N. Pathogenesis of atopic dermatitis. Clin Exp Allergy. 2015;45(3):566–74.
5. Martel BC, Lovato P, Bäumer W, Olivry T. Translational animal models of atopic dermatitis
for preclinical studies. Yale J Biol Med. 2017;90(3):389–402.
6. Elmose C, Thomsen SF. Twin studies of atopic dermatitis: interpretations and applications in
the filaggrin era. J Allergy. 2015;2015:902359.
7. Amat F, Soria A, Tallon P, Bourgoin-Heck M, Lambert N, Deschildre A, Just J. New insights
into the phenotypes of atopic dermatitis linked with allergies and asthma in children: an overview. Clin Exp Allergy. 2018;48(8):919–34.
VetBooks.ir
462
A. Diesel
8. Wilhem S, Kovalik M, Favrot C. Breed-associated phenotypes in canine atopic dermatitis. Vet
Dermatol. 2010;22:143–9.
9. Nuttal T. The genomics revolution: will canine atopic dermatitis be predictable and preventable? Vet Dermatol. 2013;24(1):10-8.e.3-4.
10. Moriello KA. Feline atopy in three littermates. Vet Dermatol. 2001;12:177–81.
11. Prost C. Les dermatoses allergiques du chat. Prat Méd Chir Anim Comp. 1993;28:151–3.
12. Roosje PJ, Thepen T, Rutten VP, et al. Immunophenotyping of the cutaneous cellular infiltrate after atopy patch testing in cats with atopic dermatitis. Vet Immunol Immunopathol.
2004;101:143–51.
13. Marsella R, Girolomoni G. Canine models of atopic dermatitis: a useful tool with untapped
potential. J Invest Dermatol. 2009;129(10):2351–7.
14. Halliwell R, the International Task Force on Canine Atopic Dermatitis. Revised nomenclature
for veterinary allergy. Vet Immuno Immunopathol. 2006;114:207–8.
15. Taglinger K, Helps CR, Day MJ, Foster AP. Measurement of serum immunoglobulin E (IgE)
specific for house dust mite antigens in normal cats and cats with allergic skin disease. Vet
Immunol Immunopathol. 2005;105:85–93.
16. Lauber B, Molitor V, Meury S, Doherr MG, Favrot C, Tengval K, et al. Total IgE and allergen-­
specific IgE and IgG antibody levels in sera of atopic dermatitis affected and non-affected
Labrador- and Golden retrievers. Vet Immunol Immunopathol. 2012;149:112–8.
17. Gilbert S, Halliwell RE. Feline immunoglobulin E: induction of antigen-specific antibody in normal cats and levels in spontaneously allergic cats. Vet Immunol Immunopathol.
1998;63:235–52.
18. Reinero CR. Feline immunoglobulin E: historical perspective, diagnostics and clinical relevance. Vet Immunol Immunopathol. 2009;132:13–20.
19. Gilbert S, Halliwell RE. Production and characterization of polyclonal antisera against feline
IgE. Vet Immunol Immunopathol. 1998;63:223–33.
20. Lee-Fowler TM, Cohn LA, DeClue AE, Spinkna CM, Ellebracht RD, Reinero CR. Comparison
of intradermal skin testing (IDST) and serum allergen-specific IgE determination in an experimental model of feline asthma. Vet Immunol Immunopathol. 2009;132:46–52.
21. Seals SL, Kearney M, Del Piero F, Hammerberg B, Pucheu-Haston CM. A study for characterization of IgE-mediated cutaneous immediate and late-phase reactions in non-allergic
domestic cats. Vet Immunol Immunopathol. 2014;159:41–9.
22. Norris Reinero CR, Decile KC, Berghaus RD, Williams KJ, Leutenegger CM, Walby WF, et al.
An experimental model of allergic asthma in cats sensitized to house dust mite or Bermuda
grass allergen. Int Arch Allergy Immunol. 2004;135:117–31.
23. Moriello KA, Stepien RL, Henik RA, Wenholz LJ. Pilot study: prevalence of positive aeroallergen reactions in 10 cats with small airway disease without concurrent skin disease. Vet
Dermatol. 2007;18:94–100.
24. Taglinger K, Day MJ, Foster AP. Characterization of inflammatory cell infiltration in feline
allergic skin disease. J Comp Pathol. 2007;137:211–23.
25. Roosje PJ, Whitaker-Menezes D, Goldschmidt MH, et al. Feline atopic dermatitis. A model for
Langerhans cell participation in disease pathogenesis. Am J Pathol. 1997;151:927–32.
26. Novak N. An update on the role of human dendritic cells in patients with atopic dermatitis. J
Allergy Clin Immunol. 2012;129:879–86.
27. Roosje PJ, Koeman JP, Thepen T, et al. Mast cells and eosinophils in feline allergic dermatitis:
a qualitative and quantitative analysis. J Comp Pathol. 2004;131:61–9.
28. Liu FT, Goodarzi H, Chen HY. IgE, mast cells, and eosinophils in atopic dermatitis. Clin Rev
Allergy Immunol. 2011;41:298–310.
29. Jarvikallio A, Naukkarinen A, Harvima IT, et al. Quantitative analysis of tryptase- and
chymase-containing mast cells in atopic dermatitis and nummular eczema. Br J Dermatol.
1997;136:871–7.
30. Beadleston DL, Roosje PJ, Goldschmidt MH. Chymase and tryptase staining of normal feline
skin and of feline cutaneous mast cell tumors. Vet Allergy Clin Immunol. 1997;5:54–8.
VetBooks.ir
Feline Atopic Syndrome: Epidemiology and Clinical Presentation
463
31. Roosje PJ, van Kooten PJ, Thepen T, Bihari IC, Rutten VP, Koeman JP, et al. Increased numbers of CD4+ and CD8+ T cells in lesional skin of cats with allergic dermatitis. Vet Pathol.
1998;35:268–73.
32. Roosje PJ, Dean GA, Willemse T, et al. Interleukin 4-producing CD4+ T cells in the skin of
cats with allergic dermatitis. Vet Pathol. 2002;39:228–33.
33. Taglinger K, Van Nguyen N, Helps CR, et al. Quantitative real-time RT-PCR measurement of
cytokine mRNA expression in the skin of normal cats and cats with allergic skin disease. Vet
Immunol Immunopathol. 2008;122:216–30.
34. Dunham S, Messamore J, Bessey L, Mahabir S, Gonzales AJ. Evaluation of circulating interleukin-­31 levels in cats with a presumptive diagnosis of allergic dermatitis. Vet Dermatol.
2018;29:284. [abstract]
35. Szczepanik MP, Wilkołek PM, Adamek ŁR, et al. The examination of biophysical parameters
of skin (transepidermal water loss, skin hydration and pH value) in different body regions of
normal cats of both sexes. J Feline Med Surg. 2011;13:224–30.
36. Szczepanik MP, Wilkołek PM, Adamek ŁR, et al. Correlation between transepidermal water
loss (TEWL) and severity of clinical symptoms in cats with atopic dermatitis. Can J Vet Res.
2018;82(4):306–11.
37. Sanford JA, Gallo RL. Functions of the skin microbiota in health and disease. Semin Immunol.
2013;25(5):370–7.
38. Rodrigues Hoffmann A, Patterson AP, Diesel A, Lawhon SD, Ly HJ, Elkins Stephenson C,
et al. The skin microbiome in healthy and allergic dogs. PLoS One. 2014;9(1):e83197.
39. Older CE, Diesel A, Patterson AP, Meason-Smith C, Johnson TJ, Mansell J, et al. The feline
skin microbiota: the bacteria inhabiting the skin of healthy and allergic cats. PLoS One.
2017;12(6):e0178555.
40. Scott DW, Miller WH, Erb HN. Feline dermatology at Cornell University: 1407 cases (1988–
2003). J Fel Med Surg. 2013;15(4):307–16.
41. Scott DW, Paradis M. A survey of canine and feline skin disorders seen in a university practice:
small animal clinic, University of Montréal, Saint-Hyacinthe, Québec (1987–1988). Can Vet J.
1990;31:830–5.
42. Hill PB, Lo A, Eden CAN, Huntley S, Morey V, Ramsey S, et al. Survey of the prevalence,
diagnosis and treatment of dermatological conditions in small animal general practice. Vet
Rec. 2006;158:533–9.
43. Griffin CE, DeBoer DJ. The ACVD task force on canine atopic dermatitis (XIV): clinical manifestations of canine atopic dermatitis. Vet Immunol Immunopathol. 2001;81(3–4):255–69.
44. Hensel P, Santoro D, Favrot C, Hill P, Griffin C. Canine atopic dermatitis: detailed guidelines
for diagnosis and allergen identification. BMC Vet Res. 2015;11:196.
45. Foster AP, Roosje PJ. Update on feline immunoglobulin E (IgE) and diagnostic recommendations for atopy. In: August JR, editor. Consultations in feline internal medicine. 5th ed. St.
Louis: Elsevier; 2006. p. 229–38.
46. Masuda K, Kurata K, Sakaguchi M, Yamashita K, Hasegawa A, Ohno K, Tsujimoto H. Seasonal
rhinitis in a cat sensitized to Japanese cedar (Cryptomeria japonica) pollen. J Vet Med Sci.
2001;63:79–81.
VetBooks.ir
Feline Atopic Syndrome: Diagnosis
Ralf S. Mueller
Abstract
Feline atopic syndrome is an aetiological diagnosis of a disease caused by environmental or dietary allergens. As such there is currently no single test reliably
differentiating feline atopic syndrome from its differential diagnoses. This syndrome is associated with a number of clinical reaction patterns such as miliary
dermatitis, eosinophilic granuloma, pruritus leading to non-inflammatory alopecia or ulcerative and crusty dermatitis. The diagnosis is confirmed by ruling out
all differential diagnoses based on history and clinical examination. Hence, the
diagnostic approach is different with the various reaction patterns. As adverse
food reaction and flea bite hypersensitivity are differential diagnoses for all these
reaction patterns, excellent ectoparasite control and an elimination diet are part
of the recommended diagnostic work-up for all cats with suspected feline atopic
syndrome. Depending on the clinical findings, other diagnostic tests such as
cytology, Wood’s lamp, trichogram, fungal culture or biopsy may be indicated.
Introduction
In contrast to canine atopic dermatitis, which has distinct clinical features, feline
atopic syndrome is characterized by a number of cutaneous reaction patterns that
look distinctively different [1, 2]. Miliary dermatitis, eosinophilic granuloma complex or pruritus without lesions, either leading to non-inflammatory alopecia or
secondary excoriations with ulceration and crusting, can all be due to feline atopic
syndrome. Similar to canine atopic dermatitis, feline atopic syndrome is a diagnosis
R. S. Mueller (*)
Centre for Clinical Veterinary Medicine, München, Germany
e-mail: dermatologie@medizinische-kleintierklinik.de
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_22
465
VetBooks.ir
466
R. S. Mueller
based on history, clinical signs and exclusion of differential diagnoses [3]. However,
each of the above-mentioned reaction patterns has a different list of differential
diagnoses and consequently needs a slightly different approach. This chapter will
discuss the differential diagnoses of the various cutaneous reaction patterns frequently associated with feline atopic syndrome as well as the diagnostic approach
for each of those.
General Principles of Diagnosis
A thorough history and clinical examination are essential for the formulation of a
list of differential diagnoses for each of the cutaneous reaction patterns seen regularly with feline atopic syndrome. Important questions to be asked depend on the
individual reaction pattern and the differential diagnoses possibly responsible for
it. Fleas, food or environmental allergens can cause all of the stated clinical signs
and thus questions about current ectoparasite control, feeding habits, faecal consistency (when owners have access to the faeces) and attempted elimination diets
are relevant for all those reaction patterns [1]. Other diagnoses are only associated
with selected patterns. For example, an infestation with Otodectes cynotis has been
reported as a cause of miliary dermatitis [4], but not with eosinophilic granuloma,
and questions about previous ear disease and other affected animals in the household are important. Non-inflammatory alopecia may be caused by demodicosis [4]
or rarely endocrine disease or alopecia areata, diseases not considered in a cat with
miliary dermatitis. The age of the patient and careful questioning with regard to
systemic signs may provide clinical clues for endocrine disease. Once the list of
differential diagnoses and their order of priority based on history and clinical findings have been established, diagnostic tests to rule out or confirm those diagnoses
are undertaken. The efforts spent to achieve a confirmed diagnosis as quickly as
possible will of course also be determined by the owner and his or her willingness
to invest time and money. In some patients, tests will be performed subsequently in
the order of disease likelihood or necessity to rule out (e.g. a dermatophyte culture
in a Persian cat), and other cat owners will choose to perform an array of tests at
the same time to rule out a number of differential diagnoses fairly quickly. Once
every other differential diagnosis has been ruled out, the diagnosis of feline atopic
syndrome is confirmed.
Approach to the Cat with Miliary Dermatitis
Although miliary dermatitis (Fig. 1) is often assumed to be caused by allergic reactions against fleas and environmental or food allergens (and indeed those are the
cause in the majority of cats), other causes are possible and may need to be considered in individual patients. Ectoparasites other than fleas, notably mites such
VetBooks.ir
Feline Atopic Syndrome: Diagnosis
467
Fig. 1 Miliary dermatitis
with small crusts in a
domestic shorthaired cat
Table 1 Diseases causing or
contributing to miliary
dermatitis
Allergies
Infectious diseases
Immune-mediated
diseases
Neoplastic diseases
Nutritional deficiencies
Flea bite hypersensitivity
Environmental atopic syndrome
Food-induced atopic syndrome
Dermatophytosis
Bacterial infection
Notoedres cati
Otodectes cynotis
Demodex cati
Pemphigus foliaceus
Mast cell tumour
Fatty acid deficiency
as Cheyletiella blakei or Otodectes cynotis, may also lead to miliary dermatitis
(Table 1). Infections with dermatophytes or bacteria may cause or contribute to
clinical signs of miliary dermatitis. On rare occasions, pemphigus foliaceus and
mast cell tumours can present with similar small crusted papules. Finally, some
nutritional deficiencies such as a lack of essential fatty acids in the diet may cause
miliary dermatitis. In most circumstances, this is unlikely with current feeding practices. Depending on the clinical history and physical examination, skin scrapings,
ectoparasite control, skin cytology or biopsy and elimination diets may be indicated
to work up individual cats with miliary dermatitis. Skin biopsy is the diagnostic test
of choice to differentiate allergic from immune-mediated or neoplastic skin disease.
However, it is less likely to be able to differentiate between ectoparasitic and allergic causes and cannot reliably differentiate causes of allergy.
468
R. S. Mueller
VetBooks.ir
Approach to the Cat with Pruritus
Initial pruritus without lesions may lead either to non-inflammatory alopecia or to
ulcerations and crusting due to self-trauma, usually in the area of the head and neck.
Those reaction patterns have different possible aetiologies.
Non-inflammatory alopecia (Fig. 2) is most frequently caused by flea, food or
environmental allergens or any combination thereof [1, 2]. A major differential diagnosis for a pruritic cat with non-inflammatory alopecia is psychogenic alopecia [5].
Major changes in the environment, such as a physical move from countryside to a
town or city, a new cat moving into the neighbourhood, a new baby or animal in the
household or changes in the work hours of the family, may cause excessive licking in some cats. Consulting a veterinary behaviourist may be helpful in such cases
and is recommended in those cases which do not respond to an elimination diet or
flea control. In early stages of dermatophytosis, associated mild scaling and a fine
papular rash may not be present or may be overlooked [6], and fungal tests such
as Wood’s lamp, trichograms, cultures or PCR may be useful diagnostic options.
However, keep in mind the possibility of false-positive PCR reactions due to transient environmental contamination. Very rarely, endocrine diseases such as hyperadrenocorticism may cause non-pruritic non-inflammatory alopecia in the cat, typically
associated with other systemic signs [7–9]. Hormonal testing is needed in such cats.
Unusual alopecias such as telogen effluvium (where a stressful event sends all hair
follicles of a certain area into a synchronized telogen (the resting phase) and alopecia occurs 6–12 weeks later when the new hair is regrowing in the deep dermis) or
anagen defluxion (where a severe metabolic disease or chemotherapy leads to the
production of damaged hair shafts that break off within the follicular lumen leading
to alopecia) can be ruled out or suspected by a thorough history. Ventral abdominal
alopecia associated with demodicosis has been diagnosed in restricted geographical
areas worldwide and associated with pruritic non-­inflammatory alopecia (Table 2).
Head and neck pruritus leading sometimes to widespread crusting and ulceration (Fig. 3) may be due to environmental allergens, but food allergens, flea
bite hypersensitivity and other ectoparasite infestations such as Notoedres cati or
Otodectes cynotis may also be considered [1, 2]. Secondary infections with bacteria or yeast occur frequently. Infections with dermatophytosis may be associated
with pruritus if the skin is inflamed. If there is a previous clinical history of an
Fig. 2 Non-inflammatory
hypotrichosis and alopecia
on the ventrum of a
7-year-old, male castrated
domestic shorthaired cat
Feline Atopic Syndrome: Diagnosis
VetBooks.ir
Table 2 Diseases causing
alopecia in the cat
469
Allergies
Psychogenic diseases
Infectious diseases
Endocrine diseases
Drug reaction
Miscellaneous diseases
Flea bite hypersensitivity
Environmental atopic syndrome
Food-induced atopic syndrome
Psychogenic alopecia
Dermatophytosis
Demodex cati
Hypothyroidism
Hyperadrenocorticism
Methimazole-induced alopecia
Telogen effluvium
Anagen defluxion
Fig. 3 A large crust and
excoriations in a cat with
severe pruritus on the head.
(Courtesy of Dr. Chiara
Noli)
upper respiratory tract infection and mucosal surfaces are affected as well, viral
infections with feline herpes- or calicivirus should be considered. On rare occasions cowpox virus infections may also lead to variably pruritic, ulcerative and
crusty skin disease with fever and anorexia [10]. Cowpox is inoculated through the
bite wound of a rodent and initially a solitary lesion develops. Fever and multifocal cutaneous lesions result from a subsequent viremia. Such cats may be highly
infectious and have been reported to die from viral pneumonia. PCR testing of
crusts is the diagnostic tool of choice, it is fast, and the poxvirus inclusions are
rich in the crust. On histopathology, cowpox virus is identified through intracytoplasmic eosinophilic inclusion bodies and herpes virus through an eosinophilic
dermatitis, folliculitis and furunculosis and with careful searching for amphophilic
intranuclear inclusion bodies.
Pruritus may be caused by flea, food or environmental allergens, but other diseases
such as medication reactions or, in older cats, paraneoplastic pruritus may also be
considered. If a hyperthyroid cat develops pruritus on methimazole, discontinuation
VetBooks.ir
470
R. S. Mueller
of the medication typically leads to a quick resolution of the self-trauma, and an alternative treatment for hyperthyroidism should be elected. In older pruritic cats, particularly with Malassezia infections of the head or neck, paraneoplastic skin disease
should be considered. Depending on history and physical examination, ultrasonography, radiography (and/or CT/MRI), lymph node aspirates, complete blood counts and
serum biochemistry may all be indicated when paraneoplastic pruritus is considered.
pproach to the Cat with Lesions of the Eosinophilic
A
Granuloma Complex
Lesions of the eosinophilic granuloma complex may be observed with feline atopic
syndrome [1, 2]. Indolent lip ulcers (Fig. 4) (often asymmetrical ulcers frequently
on the mucosal margin of the upper lip and often covered with a thick yellowish adherent exudate), eosinophilic granulomas (Fig. 5) (papular to linear lesions,
often eroded or ulcerated, frequently found on the caudal thighs) and (typically
highly pruritic) eosinophilic plaques (Fig. 6) found on the ventral abdomen and
inner thighs can all be caused by flea, food or environmental allergens, and thus
Fig. 4 Indolent ulcer on
the upper lip of a
6-year-old female domestic
shorthaired cat. (Courtesy
of Dr. Chiara Noli)
Fig. 5 A linear lesion of
eosinophilic granuloma on
the thigh. (Courtesy of Dr.
Chiara Noli)
VetBooks.ir
Feline Atopic Syndrome: Diagnosis
471
Fig. 6 Same cat as in
Fig. 4: a large eosinophilic
plaque on the abdomen.
(Courtesy of Dr. Chiara
Noli)
Fig. 7 Impression smear
from a plaque: neutrophils
with intra- and
extracytoplasmatic cocci
and rod-shaped bacteria.
The latter probably origin
from the oral flora (Diff
Quick, 1000×). (Courtesy
of Dr. Chiara Noli)
ectoparasite control and an elimination diet are part of the thorough diagnostic
work-up of any cat with lesions belonging to the eosinophilic granuloma complex.
On occasion, squamous cell carcinoma may be a differential diagnosis, particularly
in older cats with lesions in non-pigmented or sparsely haired areas of the head. In
those cats, a biopsy is also indicated.
Ruling Out Skin Infections
Although skin infections are rarer in cats than in dogs, they do occur in the feline
species and may contribute significantly to both pruritus and clinical signs. They
need to be recognized and treated to achieve optimal therapeutic outcome. A cytologic evaluation of an impression smear is the test of choice to identify bacterial
or yeast infections [11]. If the skin and crusts are very dry, a better yield is often
achieved by removing a few crusts and obtaining cytology from the underlying
surface of the crusts. Neutrophils with intracellular bacteria (Fig. 7) confirm a bacterial skin infection without a doubt. The presence of bacteria or yeast has to be
VetBooks.ir
472
R. S. Mueller
Fig. 8 Numerous
Malassezia yeasts from the
skin of an allergic cat (Diff
Quick 400×). (Courtesy of
Dr. Chiara Noli)
interpreted in light of the numbers of organisms, clinical signs and the sampling
site. Large numbers of Malassezia spp. yeasts (Fig. 8) have been reported to be a
possible clinical clue for internal malignancies in the cat; however, they can also
be found with allergic skin disease. Examination of the skin with a Wood’s lamp,
trichograms, fungal cultures or PCR for fungal antigens may be useful in patients
with possible dermatophytosis.
Ruling Out Ectoparasites
It is important to identify the type of ectoparasite control the owner conducts,
which exact product is administered, how often and to which of the animals in the
household. Some of the products on the market have a high efficacy for fleas and
ticks; others can also be used to treat mite infestations. An effective and complete
ectoparasite control should address not just fleas but also mites. Macrocyclic lactones or isoxazolines are examples of such ectoparasiticides. Whether additional
environmental control is needed in addition to regular adulticides will depend on
the individual patient, environment and climate. Flea proliferation is facilitated by
warm and humid climates. With large numbers of immature stages in a conducive
environment, spraying the house or apartment with an insect growth regulator such
as methoprene or pyriproxyfen will hasten clinical improvement in affected cats.
Similarly, such environmental control may be needed in households with multiple
animals and consequently a large environmental load of immature flea stages such
as eggs, larvae and pupae.
Performing an Elimination Diet
At this point, an elimination diet is the only reliable test to identify feline atopic
syndrome caused by food antigens [12]. This involves – theoretically – the feeding of a protein source the animal has never received before. In cats, however,
VetBooks.ir
Feline Atopic Syndrome: Diagnosis
473
that seemingly simple condition is frequently difficult to achieve. First, many cats
receive a far more varied diet than their canine counterparts, and it is not unusual
that feline patients get a different protein source every day of the week. Second,
cats are creatures of habit and more easily refuse to eat a new diet than dogs. In
addition, refusal to eat for a few days increases the risk of development of hepatic
lipidosis in the cat, and starvation until compliance is achieved is absolutely not
recommended. Consequently, several different food sources may have to be trialled before a successful elimination diet can be conducted, and the author advises
owners to have the choice of two protein sources available should the cat suddenly
decide not to eat.
Owners can choose between a home-cooked elimination diet, commercial
selected protein diets and hydrolysed diets. Many commercial selected protein
diets have been shown to be contaminated with other, off-label protein sources,
although the clinical relevance of those contaminations has not been evaluated.
Consequently, the author prefers home-cooked or extensively hydrolysed diets for
the diagnosis of food-induced feline atopic syndrome. Cats are obligate carnivores,
so that when choosing a home-cooked diet, they can be fed pure protein. A carbohydrate source is not essential and may decrease palatability and compliance in
cats. Ideally, the protein source is phylogenetically distant from the originally fed
protein. If the cat received a predominantly chicken- and turkey-based cat food,
then switching, for example, to duck may not be as suitable as rabbit or horse.
Similarly, if the cat received a beef- or lamb-based diet, then deer or goat may not
be the ideal alternative, as the chance of cross-reactivity between those allergens
is probably much higher than if ostrich or crocodile is chosen for that particular
cat. However, clinical cross-reactivities have not been established in cats with food
allergies at this point in time.
The diet should be fed exclusively for approximately 8 weeks, during which time
more than 90% of the cats with adverse food reactions will improve [13]. In those
8 weeks, no other protein sources should be permitted. A cat with access to outdoors
technically needs to be confined indoors during the entire diet. If that is not possible
and the cat does not respond, then an adverse food reaction cannot reliably be ruled
out. However, this may indeed be too stressful for the cat. In the author’s opinion, it
may be still worthwhile conducting an elimination diet in some indoor-outdoor cats,
because of the possibility that just a reduction of the amount of allergy-inducing
protein may lower the pruritic threshold. In a multi-cat household, all cats should
receive the elimination diet or the patient should be fed completely separately to
avoid unintended intake of a different protein source.
If there is no clinical improvement after 8 weeks of an appropriate elimination
diet, then an adverse food reaction is very unlikely. If however there was clinical
improvement, then a re-challenge with the previous diet is essential as this improvement may be due to the diet, but may also be due to seasonal changes, different or
more reliably administered concurrent treatments and other reasons not related to
the diet. If the re-challenge with the previous diet leads to recurrence of clinical
signs, which resolve again when the elimination diet is fed, the diagnosis of adverse
food reaction is confirmed. Long-term, the offending allergen(s) can be identified by
sequential re-challenges with individual proteins, the cat may be fed a commercial
VetBooks.ir
474
R. S. Mueller
hydrolysed or selected protein diet, or the elimination diet may be continued. If the
owner opts for the latter, it is recommended to consult a veterinary nutritionist in
order to balance the home-prepared diet and avoid nutritional deficiencies.
Conclusion
Feline atopic syndrome is an aetiological diagnosis associated with a number of
clinical reaction patterns such as miliary dermatitis, eosinophilic granuloma, pruritus leading to non-inflammatory alopecia or ulcerative and crusty dermatitis. The
diagnosis is confirmed by ruling out all differential diagnoses based on history and
clinical examination. As adverse food reaction and flea bite hypersensitivity are
differential diagnoses for all these reaction patterns, excellent ectoparasite control
and an elimination diet are part of the recommended diagnostic work-up for all cats
with suspected feline atopic syndrome. Depending on the clinical findings, other
diagnostic tests such as cytology, Wood’s lamp, trichogram, fungal culture or biopsy
may be indicated.
References
1. Hobi S, Linek M, Marignac G, Olivry T, Beco L, Nett C, et al. Clinical characteristics and
causes of pruritus in cats: a multicentre study on feline hypersensitivity-associated dermatoses.
Vet Dermatol. 2011;22:406–13.
2. Ravens PA, Xu BJ, Vogelnest LJ. Feline atopic dermatitis: a retrospective study of 45 cases
(2001–2012). Vet Dermatol. 2014;25:95–102, e27-8
3. DeBoer DJ, Hillier A. The ACVD task force on canine atopic dermatitis (XV): fundamental
concepts in clinical diagnosis. Vet Immunol Immunopathol. 2001;81:271–6.
4. Scheidt VJ. Common feline ectoparasites part 2: Notoedres cati, Demodex cati, Cheyletiella
spp. and Otodectes cynotis. Feline Pract. 1987;17:13–23.
5. Waisglass SE, Landsberg GM, Yager JA, Hall JA. Underlying medical conditions in cats with
presumptive psychogenic alopecia. J Am Vet Med Assoc. 2006;228:1705–9.
6. Scarampella F, Zanna G, Peano A, Fabbri E, Tosti A. Dermoscopic features in 12 cats with dermatophytosis and in 12 cats with self-induced alopecia due to other causes: an observational
descriptive study. Vet Dermatol. 2015;26:282–e63.
7. Boord M, Griffin C. Progesterone secreting adrenal mass in a cat with clinical signs of hyperadrenocorticism. J Am Vet Med Assoc. 1999;214:666–9.
8. Rand JS, Levine J, Best SJ, Parker W. Spontaneous adult-onset hypothyroidism in a cat. J Vet
Intern Med. 1993;7:272–6.
9. Zerbe CA, Nachreiner RF, Dunstan RW, Dalley JB. Hyperadrenocorticism in a cat. J Am Vet
Med Assoc. 1987;190:559–63.
10. Appl C, von Bomhard W, Hanczaruk M, Meyer H, Bettenay S, Mueller R. Feline cowpoxvirus infections in Germany: clinical and epidemiological aspects. Berliner und Münchner
Tierärztliche Wochenschrift. 2013;126:55–61.
11. Mueller RS, Bettenay SV. Skin scrapings and skin biopsies. In: Ettinger SJ, Feldman EC,
Cote E, editors. Textbook of veterinary internal medicine. Philadelphia: W.B. Saunders; 2017.
p. 342–5.
12. Mueller RS, Unterer S. Adverse food reactions: pathogenesis, clinical signs, diagnosis and
alternatives to elimination diets. Vet J. 2018;236:89–95.
13. Olivry T, Mueller RS, Prelaud P. Critically appraised topic on adverse food reactions of companion animals (1): duration of elimination diets. BMC Vet Res. 2015;11:225.
VetBooks.ir
Feline Atopic Syndrome: Therapy
Chiara Noli
Abstract
Feline allergic dermatitis is a chronic disease and allergen avoidance, when possible, is the best management option. If this is not possible, then a combination
of aetiologic, symptomatic, topical, antimicrobial and nutritional therapy is
implemented, depending on the individual case. Aetiologic therapy is based on
allergy test and hyposensitization, which will be curative only in a minority of
cases. All other cases will need some sort of symptomatic therapy, possibly
avoiding long-term administration of glucocorticoids. Alternative systemic treatments include ciclosporin, antihistamines, oclacitinib, palmitoylethanolamide,
maropitant and PUFAs: not all of these are effective in every case and some are
not registered for the cat. Topical treatments are not easy to apply in cats and only
a few studies confirm their efficacy. The pros and cons of allergy testing and
hyposensitization, and of topical and/or systemic symptomatic treatment will be
discussed in this chapter.
Introduction
Feline allergic dermatitis is a chronic disease. The clinician must make the client
understand that unless the offending allergen(s) are identified and removed, a cure
is rarely possible. The keys to a successful management of allergic dermatitis are
client education, long-term commitment to the treatment protocol and a combination of aetiologic, symptomatic, topical, antimicrobial and nutritional therapy. The
choice of the therapeutical plan will depend on the individual case, that is, on both
cat (severity of the lesions and temper of the patient) and owner (economical possibilities, patience, time to devote to the cat, personal preferences). The pros and cons
C. Noli (*)
Servizi Dermatologici Veterinari, Peveragno, Italy
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_23
475
VetBooks.ir
476
C. Noli
of allergy testing and hyposensitization, and of topical and/or systemic symptomatic treatment should be clearly explained, including possible combinations – and
costs – to help the owner make an informed choice. A practical guidance on how to
therapeutically approach allergic cats is offered in Box 1.
Box 1: Practical Therapeutical Approach to the Pruritic Cat
1. Diagnostic period (from first presentation to end of elimination diet):
–– Oral/topical flea control is advised in every case.
–– For non-seasonal pruritus, the cat should undergo a 2-month-long elimination diet, better if with hydrolysed food.
–– If pruritus is important and needs to be decreased, in the meanwhile the cat
can be administered short-acting oral corticosteroids at tapering doses possibly every other day for the first 6 weeks. As an alternative oclacitinib or
maropitant can be considered. At this stage ciclosporin should be avoided,
as the lag time is very long and it takes also a long time for pruritus to come
back after withdrawal. This makes it difficult to evaluate the diet.
2. First months of allergen specific immunotherapy (ASIT):
–– Oral/topical flea control is advised in every case during the whole ASIT
period.
–– If pruritus is mild to moderate, consider antihistamines and/or ultramicronized PEA, EFAs, dermatological food, and topical hydrocortisone
aceponate.
–– If these do not work or if pruritus is moderate to severe, then consider
ciclosporin or oclacitinib or maropitant during the first few months of
ASIT (induction phase). While systemic corticosteroids can be administered for a few days at the beginning of the ASIT phase (particularly if
ciclosporin is chosen as maintenance therapy), their long-term use should
be avoided, as they could possibly interfere with the desensitization mechanism of ASIT. Every 2–3 months, antipruritic therapies could be withdrawn to better evaluate ASIT efficacy.
3. Long-term symptomatic management:
–– Oral/topical flea control is advised in every case.
–– If pruritus is mild to moderate, consider antihistamines and/or ultramicronized PEA, EFAs, dermatological food, and topical hydrocortisone aceponate, together or in combination.
VetBooks.ir
Feline Atopic Syndrome: Therapy
477
–– If these do not work or if pruritus is moderate to severe, then consider long-­
term ciclosporin administration. Corticosteroids can be associated in the
first 2 weeks.
–– If ciclosporin is not an option (e.g. for g.e. upset), then alternatives are low-­
dose corticosteroids given every other day (better if associated with steroid-sparing products, such as antihistamines, EFA or ultramicronized
PEA) or oclacitinib or maropitant.
4. Management of the flare:
–– Flares are best managed with short courses (5–15 days) of high-dose corticosteroids. Long-term managements (item no. 3) should then be instituted
afterwards, if possible.
Therapy of Allergic Dermatitis and Quality of Life
Pruritus and self-induced skin lesions due to licking and scratching have a significant negative impact on the cat’s and the owner’s quality of life (QoL) [1], and
therapy aiming at decreasing discomfort should be considered from the very first
consultation. However, in two studies on the treatment of feline allergy, decrease
of pruritus and lesions was always greater than improvement of QoL [2, 3]. This is
due to the fact that administration of therapies and repeated visits to the veterinarian have a negative impact on the QoL of both cats and owners, as treating cats is
certainly more difficult and a bigger source of psychological stress than treating
dogs. This fact should be considered when designing a therapeutic plan for the
allergic feline patient, and the plan should be sustainable by the cat and owner
over a long period of time. Way of administration (oral, topical, injectable), formulation (tablets, oral liquid, lotion, spray) and frequency should be tailored to the
individual patient and owner. Feeding a “dermatological” diet and/or essential fatty
acids and/or palmitoylethanolamide (PEA) supplements mixed with food may be a
non-­traumatic way of decreasing inflammation and pruritus and the need (dose and
frequency) of other antipruritic drugs.
Aetiologic Therapy
Identification of Allergens: Allergy Testing in Cats
Allergy testing is necessary for the identification of the allergens putatively responsible for pruritus and skin lesions observed in hypersensitive cats, but it cannot be
used to diagnose allergy per se, as several healthy cats show positive results and
VetBooks.ir
478
C. Noli
some allergic ones have negative tests [4–7]. Intradermal testing as used in dogs is of
limited value in the feline species, because wheals are small, soft and transient and
tests are difficult to interpret. The use of fluorescein may improve readability and
reliability of intradermal tests [8, 9]. One problem with feline allergy skin testing is
that, until now, allergen solutions standardized for the dog’s skin were used in cats,
with limited knowledge regarding their suitability. Initial investigations have been
published on allergen threshold concentrations in cats, albeit only for pollens and in
healthy animals [10]. A further problem is that all cats need to be anaesthetized, as
stress-related cortisol release interferes with wheal formation [11]. In order to overcome these problems, percutaneous (prick) testing is currently object of investigation
in cats and is perceived to be a good alternative to intradermal testing [12, 13], but no
kit is yet commercially available specific to the feline species. As in dogs, intradermal allergy tests should not be performed in cats being treated with corticosteroids.
Serum testing is easier to perform and is offered by several laboratories over
the world. It has the advantage of being easy to carry out (just one blood sample),
does not need anaesthesia and can be performed in cats treated with corticosteroids. While in vitro allergy tests are not able to differentiate allergic from normal
cats [4–7], they can be useful for the choice of the allergens to be included in the
ASIT solution. There are no studies determining that one methodology is preferable
over another one. The frequently used and well-studied method based on the cloned
alpha chain of the human high-affinity IgE receptor (FcE-RI) (Allercept®; Heska
AG, Fribourg, Switzerland) can also be used in cats. A recent study found a strong
agreement between results of a rapid screening immunoassay (Allercept® E-Screen
2nd Generation; Heska AG, Fribourg, Switzerland) and the complete Allercept
panel; the screening assay may thus be beneficial for predicting the results of the
complete-­panel serum allergen-specific IgE assay [4].
Allergen Avoidance
Allergen avoidance is useful if the offending allergens have been correctly identified.
Cats with indoor allergies (e.g. against house dust mites such as Dermatophagoides
spp. and storage mites such as Tyrophagus, Acarus and Lepidoglyphus spp.) or danders can be allowed to spend more time outdoors. As house dust mite levels are much
higher in bedrooms than in the rest of the house, limiting the cat’s access to these
rooms may be of help. In the case of indoor allergens, frequent vacuuming with a
“high-efficiency particle air filter” (HEPA) vacuum cleaner may reduce the allergen
load, or protective furniture covers designed for human asthmatics may be of value.
Sprays or foggers (devices that produce a fine mist) containing acaricidal agents or
insect growth regulators may be helpful in cases of house dust/storage mite allergy.
Using benzoyl-benzoate sprays on a regular basis on beddings, carpets, rugs, furniture, etc. not only kills the mites but also degrades their metabolites (allergens). One
study on house dust-/storage mite-­sensitive dogs showed that the use of benzyl benzoate spray at home induces 48% resolution and 36% improvement of pruritus [14].
Unfortunately there are no studies yet on allergen avoidance in cats.
Feline Atopic Syndrome: Therapy
479
VetBooks.ir
Immunotherapy
Allergen-specific immunotherapy (ASIT) is the aetiological treatment of choice
in cases where the duration of pruritus is longer than 4 months a year. Allergens
are administered at increasing concentration and dose and at decreasing frequency,
generally by subcutaneous injection. The mechanism of action of immunotherapy
has not been investigated in cats. In dogs and humans, it seems that a shift from a
Th2- to a Th1-biased immune response and increase in T-regulatory lymphocytes is
responsible for the development of tolerance [15]. Protocols vary depending on the
producer and adjuvant. ASIT is considered safe and effective in cats, with good to
very good responses (improvement by at least 50%) achieved in 50–80% of treated
patients [16–20]. Adverse events such as increased pruritus or anaphylaxis are considered less common than in dogs [17].
As in dogs, clinical results measured by a decrease in pruritus and skin lesions
are seen anywhere from 3 to 18 months after starting treatment. Thus, during the
initial phase of immunotherapy, symptomatic treatment may be needed. If the treatment is effective, ASIT maintenance therapy is given for the rest of the patient’s
life. Only a proportion of the cases will be controlled by immunotherapy alone,
while others will require adjunctive symptomatic therapy for at least part of the
year. In a yet unpublished retrospective study conducted by the author, about 10%
of the cats could achieve remission of the allergy after 4–5 years and could withdraw ASIT without relapses. A similar observation was also reported by Vidémont
and Pin [21].
An alternative sublingual administration option is currently available, with anecdotal efficacy similar to the subcutaneous way; however there are no published
reports yet in cats. Rush immunotherapy (administering the whole induction phase
in a few hours, under medical control) was investigated in a small number of cats
and was considered safe and effective [22].
Symptomatic Therapy
Doses, administration and adverse effects of the drugs mentioned hereunder are
summarized in Table 1.
Glucocorticoids
Glucocorticoids are very effective in suppressing the signs of allergic dermatitis.
Pharmacological data on glucocorticoids in felines are scarce: cats seem to need
higher doses than dogs, as they have half the density of glucocorticoid receptors in
the skin and liver [23] and metabolize the active prednisolone better than the prodrug prednisone [24].
Usual protocols suggest to administer oral prednisolone at 1–2 mg/kg or oral
methylprednisolone 0.8–1.6 mg/kg daily until remission of pruritus (usually
Dose
Induction phase q24h:
1–2 mg/kg
0.8–1.6 mg/kg
0.1–0.2 mg/kg
0.1–0.2 mg/kg
Maintenance phase: 1/2 to
1/4 of the induction dose
q48-72 h
7 mg/kg q24h the first
month, then q48h the second
month, then twice weekly as
maintenance dose, if signs
are under control
1 mg/kg q12h
Palmitoylethanolamide 10–15 mg/kg q24h
Also in association with
glucocorticoids as sparing
agent
Maropitant
2 mg/kg q24h
Oclacitinib
(off-label use)
Ciclosporin
Oral antipruritic drug
Glucocorticoids:
Prednisolone
Methylprednisolone
Triamcinolone
Dexamethasone
None
No information available for
long-term use. Liver and heart
disease
Mild pruritus and
eosinophilic granuloma
complex
Pruritus
No information available for use
longer than 2–4 weeks
Limited information available.
Increase kidney values in some cats
in one study, not observed in
another. Close monitoring is
necessary.
None
Transitory vomit and/or diarrhoea
(24%), weight loss, gingival
hyperplasia (2%), hepatic lipidosis
(2%), systemic toxoplasmosis
Kidney disease, liver disease,
positive FIV and/or FeLV
status, malignancies, eating
raw meat, hunting and eating
the preys
No information available. As a
matter of caution the same as
ciclosporin, suspect kidney
disease
Long-term use to keep
pruritus and lesions in
remission
Quick decrease of pruritus
without the use of
glucocorticoids
Side effects
Skin fragility syndrome, diabetes
mellitus, congestive heart failure,
polyuria and polydipsia, increased
susceptibility to bladder and skin
infections, demodicosis and
dermatophytosis
Contraindications
Diabetes, kidney disease, liver
disease, positive FIV and/or
FeLV status
Indication
Quick decrease of pruritus
and inflammation,
resolution of lesions of
the eosinophilic
granuloma complex
Table 1 Main antipruritic and anti-inflammatory drugs used for feline allergic dermatitis. Antihistamines are reported in Table 2
VetBooks.ir
480
C. Noli
VetBooks.ir
Feline Atopic Syndrome: Therapy
481
3–15 days), and then the dose is reduced to every other day and then further reduced
every week to the lowest dose that will control the clinical signs (generally 0.5–
0.1 mg/kg every other day). If prednisolone or methylprednisolone do not seem to
be effective, a good alternative in cats is oral dexamethasone or triamcinolone (both
at 0.1–0.2 mg/kg), which should then be tapered to 0.02–0.05 mg/kg every second
to third day for maintenance therapy. The use of methylprednisolone and triamcinolone at the doses mentioned above did not cause an increase of fructosamine
above the reference range, while triamcinolone caused a higher increase of amylase
compared to methylprednisolone [25].
The use of repositol methylprednisolone acetate (usually 15–20 mg/cat, SC),
whose duration of action ranges from 3 to 6 weeks, should be considered only for
refractory cats, when oral administration is not possible. Repeated repositol injections appear to become less and less effective with time, so that increased frequency
and/or higher doses may become necessary, with increased risks of adverse effects
development. In these cases, alternative therapies (such as oral or injectable ciclosporin) should be considered.
Cats are usually considered to tolerate glucocorticoids well; however adverse
effects can occur and can be severe [26]. Among these there are cutaneous atrophy
with skin fragility (Fig. 1), congestive heart failure, increased susceptibility to diabetes mellitus (particularly in obese cats), polydipsia and polyuria and increased
susceptibility to bladder and skin infections, including development of dermatophytosis and demodicosis. A recent study, however, found no evidence of bacteriuria in
cats treated with long-term oral or repositol glucocorticoids [27].
Hydrocortisone aceponate topical spray is useful to treat localized pruritus and
reduce the need for systemic medication. This product has been proven to cause
minimal thinning of the skin and local immunosuppression and has very low systemic absorption in dogs. An open pilot trial on ten cats determined that it is able
to improve pruritus and skin lesions in allergic felines and keep them under control
with daily or every other day maintenance administration [28].
Fig. 1 Large ulceration
due to skin fragility in a cat
being treated with 20 mg/
cat injectable
methylprogesterone acetate
once monthly for 5
consecutive months. The
cat has completely
recovered after withdrawal
of the drug
482
C. Noli
VetBooks.ir
Ciclosporin
Ciclosporin is a polypeptide derived from the fungus Tolypocladium inflatum. Its
mode of action is by inhibition of calcineurin. It has a variety of immunological
effects on multiple components of the skin immune system and is active in the acute
and chronic phase of allergic dermatitis. Ciclosporin has the same efficacy as prednisolone in the control of clinical signs of allergic dermatitis in cats [29]. Significant
reduction in pruritus should be expected in 75–85% of cases within 1 month of
treatment [30]. The initial oral dose in cats is 7 mg/kg/day [31]. This dose should
be administered for at least 1 month before, if effective, tapering it to every other
day. After another month of successful every other day administration, tapering to
twice weekly can be tried. About 15% and 60% of cats with skin allergy can be kept
under control, respectively, with every other day or twice weekly administration
[32, 33]. A lag period of about 2–3 weeks, in which no response is seen, occurs after
ciclosporin treatment is started, and owners should be warned about this. In dogs,
association of 3 weeks of prednisone or oclacitinib with ciclosporin, to quickly
decrease pruritus during the lag phase, has been described, [34, 35], but no such data
are available for the feline species.
The proprietary feline product (Atopica® for Cats, Elanco) is a microemulsified ciclosporin liquid formulation (100 mg/ml), which cannot be mixed with
water. To maximize absorption, ciclosporin should be administered 2 hours
before a meal; however recent data suggested that giving ciclosporin with food
does not alter clinical outcomes [36]. This formulation is not always palatable
when mixed with food, and when administered directly in the mouth it can cause
hypersalivation in some subjects. A syringe of fresh water may be dispensed after
ciclosporin administration in order to overcome this problem. The successful use
of injectable ciclosporin (50 mg/ml) at the dose of 2.5–5 mg/kg every 24–72 h
was recently described [37] and could be considered for the treatment of refractory cats.
Ciclosporin is usually well tolerated by cats. Reported adverse effects are
transitory vomiting and/or diarrhoea in up to one fourth of the cases, so that the
owners should be warned about their possible occurrence [38]. The co-administration of maropitant (2 mg/kg) with ciclosporin during the first 2–3 weeks
has been anecdotally suggested to decrease vomiting and provide a quick relief
of pruritus (see later for anti-pruritic effects of maropitant). Other described
adverse effects are weight loss (16%) and rarely gingival hyperplasia (Fig. 2),
anorexia and hepatic lipidosis (each 2% of the cases) [38]. Cats should be FIVFeLV negative and should not be allowed to hunt and eat raw meat, due to the
risk of developing fatal toxoplasmosis [39]. Preventive or concurrent (during
therapy) measurement of IgG and/or IgM anti-Toxoplasma serum titres does not
seem to be useful to predict the development of toxoplasmosis. Clinicians should
be alerted by the development of any neurologic and/or respiratory sign or significant weight loss (over 20%) in cats treated with ciclosporin.
VetBooks.ir
Feline Atopic Syndrome: Therapy
483
Fig. 2 Gingival
hyperplasia in a cat being
treated with daily
ciclosporin at 10 mg/kg for
3 months. The lesions
greatly improved when the
dose was lowered to 5 mg/
kg every other day
Table 2 Oral antihistamines reported to be used in allergic cats against pruritus
Antihistamine
Amitriptyline
Cetirizine
Chlorpheniramine
Clemastine
Cyproheptadine
Dose
5–10 mg/cat q12-24 h
1 mg/kg or 5 mg/cat q24h
2–4 mg/cat q8-24h
0.25–0.68 mg/cat q12h
2 mg/cat q12h
Diphenhydramine 1–2 mg/kg or 2–4 mg/cat
q8-12h
Fexofenadine
2 mg/kg up to 30–60 mg/cat q24h
Hydroxyzine
5–10 mg/cat q8-12h
Oxatomide
15–30 mg/cat q12h
Promethazine
5 mg/cat q24h
Side effects
Sleepiness
Sleepiness, soft stools
Sleepiness, vomit,
behavioural disturbances
Reported
efficacy
(% of cats
controlled)
Up to 41%
Up to 73%
Up to 50%
Up to 40%
Behavioural disturbances
Up to 50%
Antihistamines
Antihistamines inhibit the action of histamine by competitively blocking H1 receptors. As in dogs, the response to antihistamine therapy is variable, and it may be
necessary to try several different agents for a period of 15 days each to determine
which, if any, is more effective. The efficacy in terms of percentage of animals
responding to antihistamines in cats is reported (in old, uncontrolled studies, summarized by Scott 1999 [40]) to be between 20% and 73% (Table 2).
VetBooks.ir
484
C. Noli
In particular, cetirizine has been object of recent investigations in cats.
Pharmacological studies determined that cetirizine is orally well absorbed in cats
and is able to maintain high plasma concentrations for at least 24 h [41]. In an open
study, 5 mg/cat q24h determined a reduction in pruritus in 41% (13/32) of allergic
cats; however only a minority (1/13) of these improved more than 50%, while the
majority (10/13) improved less than 25% [42]. A subsequent randomized, double-­
blinded, placebo-controlled, crossover trial on the use of cetirizine 1 mg/kg q24h
in 21 allergic cats confirmed that only 10% of cats improved by more than 50%
with cetirizine versus 20% of placebo-treated patients, with no statistical difference
between groups for pruritus or lesions [43].
Oclacitinib
Oclacitinib (Apoquel®, Zoetis) is a JAK1 inhibitor registered for dogs, able to block
intracellular metabolic pathways leading to the allergic activation of inflammatory cells
and keratinocytes and to the elicitation of pruritus in neural fibres. Recently, the offlabel use of oclacitinib in cats was investigated in a pilot [44] and in a methylprednisolone-controlled study [3]. Oclacitinib given at 1 mg/kg every 12 h has an efficacy
similar to that of methylprednisolone given at the same dose, albeit with no obvious
advantage. Given for 1 month, it was generally well tolerated; however mild increases
in kidney values were observed in some cats [3]. In another study no clinical, hematological or biochemical alterations were observed in cats taking oclacitinib 1 or 2 mg/kg
twice daily for 28 days [45]. Oclacitinib could be a useful alternative treatment when
glucocorticoids are contraindicated and a rapid relief of pruritus is required. Readers
should be warned that oclacitinib is not registered in cats and that its long-term safety
is not known in this species. Regular haematology and biochemistry monitoring are
advised for long-term maintenance therapies.
Palmitoylethanolamide (PEA)
Palmitoylethanolamide (PEA) is a natural-occurring bioactive lipid present in both
animals and plants. PEA is produced by several different cell types in response to
tissue damage and acts by controlling the functionality of mast cells (it inhibits
degranulation) and other inflammatory cells such as macrophages and keratinocytes.
Consequently, PEA decreases skin inflammation and nerve sensitization in animals
with allergic dermatitis. An open pilot study on 17 cats with eosinophilic granuloma
and eosinophilic plaque showed that PEA (10 mg/kg q24h for 30 days) improved
pruritus, erythema and alopecia in 64.3% of cats and reduced the extent and severity
of eosinophilic plaques and granulomas in 66.7% [46]. Recently a product containing ultramicronized PEA (PEA-um) with improved bioavailability and efficacy was
released to the international veterinary market. A multicentre, placebo-controlled,
randomized trial determined a glucocorticoid sparing effect of PEA-um (15 mg/kg
q24h) in cats with non-seasonal allergic dermatitis [47]. In the same study, PEA-um
was showed to be able to prolong the effects of a short course of oral glucocorticoids
with virtually no significant adverse effects.
Feline Atopic Syndrome: Therapy
485
VetBooks.ir
Maropitant
Maropitant is a neurokinin-1 receptor antagonist, able to block the interaction of
substance P, a pruritogenic neurokinin, to its receptor. In an open pilot study at the
dose of 2 mg/kg, it has been reported to be effective against pruritus and lesions in
11/12 allergic cats [48]. Maropitant was well tolerated if administered once daily
for 2–4 weeks. There is no information about its safety for a long-term treatment.
Omega-3 and Omega-6 Fatty Acid Supplementation
There are only few old and uncontrolled studies investigating the efficacy of essential fatty acids (EFA) in miliary dermatitis and lesions of eosinophilic granuloma
in cats [49–52]. These publications reported efficacy in 40–60% of treated animals.
A lag period of 6–12 weeks occurs, before any benefits are seen. Probably, only a
small minority of patients can be controlled with fatty acid therapy alone. EFA may
have glucocorticoid- or ciclosporin-sparing effects, as determined in dogs, but no
studies were conducted in cats to confirm this. Feeding a good dermatological diet
may be an effective way of supplementing EFAs in allergic cats.
References
1. Noli C, Borio S, Varina A, et al. Development and validation of a questionnaire to evaluate
the quality of life of cats with skin disease and their owners, and its use in 185 cats with skin
disease. Vet Dermatol. 2016;27:247–e58.
2. Noli C, Ortalda C, Galzerano M. L’utilizzo della ciclosporina in formulazione liquida (Atoplus
gatto®) nel trattamento delle malattie allergiche feline. Veterinaria (Cremona). 2014;28:15–22.
3. Noli C, Matricoti I, Schievano C. A double-blinded, randomized, methylprednisolone controlled study on the efficacy of oclacitinib in the management of pruritus in cats with nonflea
nonfood induced hypersensitivity dermatitis. Vet Dermatol. 2019;30:110–e30.
4. Diesel A, DeBoer DJ. Serum allergen-specific immunoglobulin E in atopic and healthy cats:
comparison of a rapid screening immunoassay and complete-panel analysis. Vet Dermatol.
2011;22:39–45.
5. Bexley J, Hogg JE, Hammerberg B, et al. Levels of house dust mite-specific serum immunoglobulin E (IgE) in different cat populations using a monoclonal based anti-IgE enzyme-linked
immunosorbent assay. Vet Dermatol. 2009;20:562–8.
6. Gilbert S, Halliwell REW. Feline immunoglobulin E: induction of antigen-specific antibody in normal cats and levels in spontaneously allergic cats. Vet Immunol Immunopathol.
1998;63:235–52.
7. Taglinger K, Helps CR, Day MJ, et al. Measurement of serum immunoglobulin E (IgE) specific
for house dust mite antigens in normal cats and cats with allergic skin disease. Vet Immunol
Immunopathol. 2005;105:85–93.
8. Kadoya-Minegishi M, Park SJ, Sekiguchi M, et al. The use of fluorescein as a contrast medium
to enhance intradermal skin tests in cats. Austr Vet J. 2002;80:702–3.
9. Schleifer SG, Willemse T. Evaluation of skin test reactivity to environmental allergens in
healthy cats and cats with atopic dermatitis. Am J Vet Res. 2003;64:773–8.
10. Scholz FM, Burrows AK, Griffin CE, Muse R. Determination of threshold concentrations of
plant pollens in intradermal testing using fluorescein in clinically healthy nonallergic cats. Vet
Dermatol. 2017;28:351–e78.
VetBooks.ir
486
C. Noli
11. Willemse T, Vroom MW, Mol JA, Rijnberk A. Changes in plasma cortisol, corticotropin,
and alpha-melanocyte-stimulating hormone concentrations in cats before and after physical
restraint and intradermal testing. Am J Vet Res. 1993;54:69–72.
12. Rossi MA, Messinger L, Olivry T, Hoontrakoon R. A pilot study of the validation of percutaneous testing in cats. Vet Dermatol. 2013 Oct;24:488–e115.
13. Gentry CM, Messinger L. Comparison of intradermal and percutaneous testing to histamine,
saline and nine allergens in healthy adult cats. Vet Dermatol. 2016;27:370–e92.
14. Swinnen C, Vroom M. The clinical effect of environmental control of house dust mites in 60
house dust mite-sensitive dogs. Vet Dermatol. 2004;15:31–6.
15. Mueller RS, Jensen-Jarolim E, Roth Walter F, et al. Allergen immunotherapy in people, dogs,
cats and horses – differences, similarities and research needs. Allergy. 2018;. early view
online;73:1989. https://doi.org/10.1111/all.13464.
16. Carlotti D, Prost C. L’atopie féline. Le Point Vétérinaire. 1988;20:777–84.
17. Trimmer AM, Griffin CE, Rosenkrantz WS. Feline immunotherapy. Clin Techniques Small An
Pract. 2006;21:157–61.
18. Ravens PA, Xu BJ, Vogelnest LJ. Feline atopic dermatitis: a retrospective study of 45 cases
(2001-2012). Vet Dermatol. 2014;25:95–102.
19. Reedy LM. Results of allergy testing and hyposensitization in selected feline skin diseases. J
Am Anim Hosp Assoc. 1982;18:618–23.
20. Löewenstein C, Mueller RS. A review of allergen-specific immunotherapy in human and veterinary medicine. Vet Dermatol. 2009;20:84–98.
21. Vidémont E, Pin D. How to treat atopy in cats? Eur J Comp An Pract. 2009;19:276–82.
22. Trimmer AM, Griffin CE, Boord MJ, et al. Rush allergen specific immunotherapy protocol in
feline atopic dermatitis: a pilot study of four cats. Vet Dermatol. 2005;16:324–9.
23. Broek AHM, Stafford WL. Epidermal and hepatic glucocorticoid receptors in cats and dogs.
Res Vet Sci. 1992;52:312–5.
24. Graham-Mize CA, Rosser EJ, Hauptman J. Absorption, bioavailability and activity of prednisone and prednisolone in cats. In: Hiller A, Foster AP, Kwochka KW, editors. Advances in
veterinary dermatology, vol. 5. Oxford: Blackwell; 2005. p. 152–8.
25. Ganz EC, Griffin CE, Keys DA, et al. Evaluation of methylprednisolone and triamcinolone for
the induction and maintenance treatment of pruritus in allergic cats: a double-blinded, randomized, prospective study. Vet Dermatol. 2012;23:387–e72.
26. Lowe AD, Campbell KL, Graves T. Glucocorticoids in the cat. Vet Dermatol. 2008;19:340–7.
27. Lockwood SL, Schick AE, Lewis TP, Newton H. Investigation of subclinical bacteriuria in
cats with dermatological disease receiving long-term glucocorticoids and/or ciclosporin. Vet
Dermatol. 2018;29:25–e12.
28. Schmidt V, Buckley LM, McEwan NA, Rème CA, Nuttall TJ. Efficacy of a 0.0584% hydrocortisone aceponate spray in presumed feline allergic dermatitis: an open label pilot study. Vet
Dermatol 2012; 23: 11–6, e3–4.
29. Wisselink MA, Willemse T. The efficacy of cyclosporine a in cats with presumed atopic dermatitis: a double blind, randomized prednisolone-controlled study. Vet J. 2009;180:55–9.
30. King S, Favrot C, Messinger L, et al. A randomized double-blinded placebo-controlled study
to evaluate an effective ciclosporin dose for the treatment of feline hypersensitivity dermatitis.
Vet Dermatol. 2012;23:440–e84.
31. Roberts ES, Speranza C, Friberg C, et al. Confirmatory field study for the evaluation of ciclosporin at a target dose of 7.0 mg/kg (3.2 mg/lb) in the control of feline hypersensitivity dermatitis. J Feline Med Surg. 2016;18:889–97.
32. Steffan J, Roberts E, Cannon A, et al. Dose tapering for ciclosporin in cats with nonflea-­
induced hypersensitivity dermatitis. Vet Dermatol. 2013;24:315–22.
33. Roberts ES, Tapp T, Trimmer A, et al. Clinical efficacy and safety following dose tapering of
ciclosporin in cats with hypersensitivity dermatitis. J Feline Med Surg. 2016;18:898–905.
34. Panteri A, Strehlau G, Helbig R, et al. Repeated oral dose tolerance in dogs treated concomitantly with ciclosporin and oclacitinib for three weeks. Vet Dermatol. 2016;27:22–e7.
VetBooks.ir
Feline Atopic Syndrome: Therapy
487
35. Dip R, Carmichael J, Letellier I, et al. Concurrent short-term use of prednisolone with cyclosporine A accelerates pruritus reduction and improvement in clinical scoring in dogs with
atopic dermatitis. BMC Vet Res. 2013;3(9):173.
36. Steffan J, King S, Seewald W. Ciclosporin efficacy in the treatment of feline hypersensitivity dermatitis is not influenced by the feeding status. Vet Dermatol. 2012;23(suppl. 1):64–5.
(abstract)
37. Koch SN, Torres SMF, Diaz S, et al. Subcutaneous administration of ciclosporin in 11 allergic
cats – a pilot open-label uncontrolled clinical trial. Vet Dermatol. 2018;29:107–e43.
38. Heinrich NA, McKeever PJ, Eisenschenk MC. Adverse events in 50 cats with allergic dermatitis receiving ciclosporin. Vet Dermatol. 2011;22:511–20.
39. Last RD, Suzuki Y, Manning T. A case of fatal systemic toxoplasmosis in a cat being treated
with cyclosporin a for feline atopy. Vet Dermatol. 2004;15:194–8.
40. Scott DW, Miller WH Jr. Antihistamines in the management of allergic pruritus in dogs and
cats. J Small Anim Pract. 1999;40:359–64.
41. Papich MG, Schooley EK, Reinero CR. Pharmacokinetics of cetirizine in healthy cats. Am J
Vet Res. 2008;69:670–4.
42. Griffin JS, Scott DW, Miller WH Jr, et al. An open clinical trial on the efficacy of cetirizine
hydrochloride in the management of allergic pruritus in cats. Can Vet J. 2012;53:47–50.
43. Wildermuth K, Zabel S, Rosychuk RA. The efficacy of cetirizine hydrochloride on the pruritus of cats with atopic dermatitis: a randomized, double-blind, placebo-controlled, crossover
study. Vet Dermatol. 2013;24:576–681, e137-8.
44. Ortalda C, Noli C, Colombo S, Borio S. Oclacitinib in feline nonflea-, nonfood-induced
hypersensitivity dermatitis: results of a small prospective pilot study of client-owned cats. Vet
Dermatol. 2015;26:235–e52.
45. Lopes NL, Campos DR, Machado MA, Alves MSR, de Souza MSG, da Veiga CCP, Merlo A,
Scott FB, Fernandes JI. A blinded, randomized, placebo-controlled trial of the safety of oclacitinib in cats. BMC Vet Res. 2019;15(1):137.
46. Scarampella F, Abramo F, Noli C. Clinical and histological evaluation of an analogue of palmitoylethanolamide, PLR 120 (comicronized Palmidrol INN) in cats with eosinophilic granuloma and eosinophilic plaque: a pilot study. Vet Dermatol. 2001 Feb;12(1):29–39.
47. Noli C, Della Valle MF, Miolo A, Medori C, Schievano C; Skinalia Clinical Research Group.
Effect of dietary supplementation with ultramicronized palmitoylethanolamide in maintaining
remission in cats with nonflea hypersensitivity dermatitis: a double-blind, multicentre, randomized, placebo-controlled study.Vet Dermatol. 2019;30:387–e117.
48. Maina E, Fontaine J. Use of maropitant for the control of pruritus in non-flea, non-foodinduced feline hypersensitivity dermatitis: an open label uncontrolled pilot study. J Feline
Med Surg. 2019;21:967–72.
49. Harvey RG. Management of feline miliary dermatitis by supplementing the diet with essential
fatty acids. Vet Rec. 1991;128:326–9.
50. Harvey RG. The effect of varying proportions of evening primrose oil and fish oil on cats with
crusting dermatosis (miliary dermatitis). Vet Rec. 1993a;133:208–11.
51. Harvey RG. A comparison of evening primrose oil and sunflower oil for the management of
papulocrustous dermatitis in cats. Vet Rec. 1993b;133:571–3.
52. Miller WH, Scott DW, Wellington JR. Efficacy of DVM Derm caps liquid in the management
of allergic and inflammatory dermatoses of the cat. JAAHA. 1993;29:37–40.
VetBooks.ir
Mosquito-byte Hypersensitivity
Ken Mason
Abstract
Feline mosquito bite allergy has a worldwide distribution occurring where
cats are seasonally exposed to mosquitoes. The distinctive skin lesions are
punctate ulcers, crusts and pigmentary changes on the face, ears and nose.
Associated pruritus causes face and nose pawing resulting in bleeding. Foot
pad hyperkeratosis, crusts and pigmentary changes occur in some cats.
Confining the cat inside a screened area and in late afternoon reduces severity
of signs; intermittent corticosteroid with confinement also helps. Newer repellent pyrethroid/pyrethrins safe for cats are becoming available and prove useful to affected cats.
Introduction
Feline mosquito bite hypersensitivity is an uncommon, seasonal, visually distinctive pruritic dermatitis typically affecting the face, ears and footpads [1–4].
The disease was originally described in 1984 by Wilkinson and Bate as a seasonal variant of the eosinophilic granuloma complex that improved on hospitalization [5].
In 1991, Mason and Evans hypothesized that the cause was a mosquito bite
hypersensitivity, when they realized that lesions were restricted to short-haired
or non-haired areas, such as the nose and footpads [1]. The authors demonstrated
that clipping the hair short on the forehead resulted in lesions, when the cat was
K. Mason (*)
Specialist Veterinary Dermatologist, Animal Allergy & Dermatology Service,
Slacks Creek, QLD, Australia
e-mail: ken@dermcare.com.au
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_24
489
VetBooks.ir
490
K. Mason
Fig. 1 At examination the
cat presented with crusted
ulceration on ear tips,
erythema, crusts and
depigmentation of the
nasal bridge and paw and
small punctate ulcers and
depigmentation of the
nasal planum
Fig. 2 After 1 week of
hospitalization, the cat
presents improvement of
lesions. The forehead coat
was clipped before
returning to home
environment to prove that
areas with short coat are
predisposed to lesions
exposed to the home environment. Only some cats in a multi-cat household
developed lesions, further supporting an environmental hypersensitivity cause.
The final proof that mosquito bite caused the skin disease is demonstrated in the
sequence of photos in Figs. 1, 2, 3 and 4.
Pathogenesis and Epidemiology
Similarly to flea allergy dermatitis (FAD), mosquito bite allergy is an IgE-mediated
type I (immediate) hypersensitivity reaction [1, 2, 4]. The disease is seasonal, occurring intermittently in the spring and continuing through summer, waning in autumn
and usually absent in winter. In subsequent years, the allergy and lesion severity
may increase depending on weather pattern, favouring or not mosquito breeding.
There is no age or sex predilection; usually affected cats are adult and have been
VetBooks.ir
Mosquito-byte Hypersensitivity
491
Fig. 3 Re-examination
after 1 week at home
environment: clipped areas
present new lesions and
previously improved
lesions have flared up. The
cat was then returned home
and kept in a mosquito
netting-covered cage
outside
Fig. 4 When living at
home in an outside
environment in a mosquito
proof netting-enclosed
cage, lesions improved
again. After a hole cut in
the net, mosquitoes could
enter and bite the cat again
exposed to more than one mosquito season. Occurrence is more commonly reported
in cats that venture outside in geographic regions where mosquitoes are endemic.
In multi-cat households, only one or a few cats are affected. The disease occurs
anywhere cats are exposed to mosquitoes.
Clinical Signs
The striking and typical clinical manifestations of mosquito bite hypersensitivity
are erythema, crusted and ulcerated ear margins, papules to small nodules with focal
crusting on the haired ear surface, punctate ulcers to severely crusted lesions on
the nasal bridge and erythema, ulceration and depigmentation of the nasal planum
(Figs. 1 and 5). On the footpads, there may be hyperkeratosis often affecting the
margins and variable pigmentation alteration. Pruritus can be intense during active
mosquito challenge, leading to self-trauma and bleeding.
VetBooks.ir
492
K. Mason
Fig. 5 After the mosquitoes
bit the nose, the skin
redeveloped inflammation,
and skin biopsies were
sampled again; sutures are
visible
Fig. 6 Eosinophilic ulcer
(indolent ulcer) eroding
upper lip due to mosquito
bite hypersensitivity
There are a variety of other less typical lesions, especially in cats living in
severely mosquito-infested areas like swamps and irrigation areas, such as eosinophilic plaques, indolent lip ulcers (Fig. 6), hairless chin nodules and linear granulomas on the body. Eosinophilic keratoconjunctivitis is occasionally present and
waxes and wanes with mosquito challenge.
Regional lymph nodes, particularly the submandibular ones, may be enlarged
and the temperature may be slightly raised.
Differential Diagnoses and Diagnostic Tests
The clinical features are sufficiently characteristic to make the diagnosis in typical cases; however there are potential alternative diagnoses such as squamous cell
carcinoma and herpesvirus (FeHV-1) dermatitis, so that confirmation tests may be
needed. Herpesvirus dermatitis can present with large crusts on the nasal bridge,
and in squamous cell carcinoma, erosive, crusted lesions on the ear tip and nose may
VetBooks.ir
Mosquito-byte Hypersensitivity
493
Fig. 7 Photomicrograph
histopathology section
H&E stain showing
follicular necrosis (arrow)
and dermal inflammation
of eosinophils and
macrophages (star)
develop, particularly in white skin. Footpad hyperkeratosis can present a diagnostic
dilemma, given the difficulty of making a diagnosis when confronted with discrete
pad keratoses. In case of lesions of the eosinophilic granuloma complex, other allergic causes should be considered.
Intradermal skin test and blood immunoassay may be supportive if mosquito antigen is available. Blood haematology may show a raised eosinophil count. Similarly,
cytology of lesions and lymph nodes can be supportive if dominated by eosinophils
and may help to rule out alternative diseases, such as squamous cell carcinoma.
The diagnosis may be confirmed if isolation in a hospital or mosquito-free home
or enclosure results in resolution of acute signs within days and when return to
home outside causes a relapse of pruritus and lesions.
Histopathological examination of lesions is classically characterized by eosinophilic follicular necrosis. Common findings are eosinophilic folliculitis and furunculosis, surface serocellular crusts, hyperplastic spongiotic epidermis with eosinophil
exocytosis and micro-pustules and a diffuse dermal eosinophilic inflammation, with
a few lymphocytes and occasional flame figures (Fig. 7).
Treatment
Avoidance of mosquitoes as much as possible is the mainstay of treatment. Affected
cats should be kept indoors behind insect screened enclosures, where mosquito
exposure is prevented. Insect repellents designed for dogs or humans are toxic to
cats [6]. However, natural pyrethrin from the chrysanthemum flowers and the newer
synthetic flumethrin are safe for cats and could help to manage mosquito bite allergic patients. Very few products are approved for this disease in cats, but some, such
as collars, have proven repellent activity against sandfly vectors of leishmaniosis
[7–9]. Supplementary anti-pruritic glucocorticoids help in case of disease flares.
VetBooks.ir
494
K. Mason
Yard environmental mosquito control may be helpful and is an important preventative human health consideration. Standing water should be eliminated to decrease
mosquito breeding grounds.
Conclusion
The typical presentation of mosquito bite allergy is sufficiently distinctive to make
a diagnosis without supportive tests. However, the less typical forms and footpad
hyperkeratosis present a diagnostic challenge, and the aetiology can be overlooked,
leading to chronic high doses of corticosteroids and subsequent major adverse
effects. Keeping the cat inside in the late afternoon and overnight and flumethrin
collars are likely to be beneficial.
References
1. Mason KV, Evans AG. Mosquito bite-caused eosinophilic dermatitis in cats. J Am Vet Med
Assoc. 1991;198(12):2086–8.
2. Nagata M, Ishida T. Cutaneous reactivity to mosquito bites and its antigens in cats. Vet
Dermatol. 1997;8(1):19–26.
3. Johnstone AC, Graham DG, Andersen HJ. A seasonal eosinophilic dermatitis in cats. N Z Vet
J. 1992;40(4):168–72.
4. Ihrke PJ, Gross TL. Conference in dermatology—no. 2 mosquito-bite hypersensitivity in a cat.
Vet Dermatol. 1994;5(1):33–6.
5. Wilkinson GT, Bate MJ. A possible further clinical manifestation of the feline eosinophilic
granuloma complex. J Am Anim Hosp Assoc. 1984;20:325–31.
6. Dymond NL, Swift IM. Permethrin toxicity in cats: a retrospective study of 20 cases. Aust Vet
J. 2008;86(6):219–23.
7. Stanneck D, Kruedewagen EM, Fourie JJ, Horak IG, Davis W, Krieger KJ. Efficacy of an imidacloprid/flumethrin collar against fleas and ticks on cats. Parasit Vectors. 2012;5:82.
8. Stanneck D, Rass J, Radeloff I, Kruedewagen E, Le Sueur C, Hellmann K, Krieger K. Evaluation
of the long-term efficacy and safety of an imidacloprid 10% / flumethrin 4.5% polymer matrix
collar (Seresto (R)) in dogs and cats naturally infested with fleas and/or ticks in multicentre
clinical field studies in Europe. Parasit Vectors. 2012;5:66.
9. Brianti E, Falsone L, Napoli E, et al. Prevention of feline leishmaniosis with an imidacloprid
10%/flumethrin 4.5% polymer matrix collar. Parasit Vectors. 2017;10:334.
VetBooks.ir
Autoimmune Diseases
Petra Bizikova
Abstract
Autoimmune skin diseases (AISDs) in cats are very rare and account for less
than 2% of all skin diseases for which cats are seen by dermatologists. The most
common AISD seen in this species is pemphigus foliaceus, for which numerous
case reports and case series can be found in the literature. In contrast, other
AISDs are very rare and limited to only few case reports published in the peer-­
reviewed literature over the last two decades. Many of these diseases are clinically and histologically homologous to diseases described in people and dogs,
and, although the pathomechanism of these feline counterparts is unknown, similar mechanisms leading to the disruption of the epidermal cohesion or destruction of skin adnexae are hypothesized. Such mechanisms involve autoantibodies
in diseases like pemphigus foliaceus, pemphigus vulgaris, paraneoplastic pemphigus, and autoimmune subepidermal blistering diseases, or autoreactive T cells
in diseases like paraneoplastic pemphigus, cutaneous lupus, and vitiligo. This
chapter will provide an overview of the current knowledge about feline AISDs
available in the published literature.
Introduction
A healthy immune system protects the body from an onslaught of invading pathogens as well as from its own damaged or potentially neoplastic cells on a daily basis.
Under specific circumstances (genetics, environment, infection, etc.), however, the
same immune system may get awry and start targeting self-antigens. This break of
P. Bizikova (*)
North Carolina State University, College of Veterinary Medicine, Raleigh, NC, USA
e-mail: pbiziko@ncsu.edu
© Springer Nature Switzerland AG 2020
C. Noli, S. Colombo (eds.), Feline Dermatology,
https://doi.org/10.1007/978-3-030-29836-4_25
495
VetBooks.ir
496
P. Bizikova
a self-tolerance results in an injury to the body, which at the turn of the twentieth
century was named a horror autotoxicus by Paul Ehrlich. Such autoimmune attack
can be caused by autoantibodies (e.g., pemphigus) or by autoreactive T lymphocytes (e.g., cutaneous lupus). Autoimmune skin diseases (AISDs) are rare in cats
and account for less than 2% of all skin diseases for which cats are seen by a dermatologist [1]. The most common AISD seen in this species is pemphigus foliaceus,
for which numerous case reports and case series can be found in the literature. In
contrast, other AISDs are very rare and limited to only few case reports published in
the peer-reviewed literature over the last two decades. Due to the rarity of AISDs in
cats, information about the identity of the autoantigen and the disease pathogenesis
remains unknown.
utoimmune Skin Diseases Affecting the Epidermal
A
and Dermo-Epidermal Adhesion
An intact skin is a critically important organ that functions as a first-line defense
mechanism against physical and chemical damage. Its integrity is dependent on
complex structures maintaining cell-cell and cell-matrix adhesions [2, 3]. Several
AISDs disrupting this cohesion have been recognized in cats. The mechanism by
which this adhesion is disrupted varies depending on the type of disease.
(a) Disruption of keratinocyte adhesion – intra-epidermal blister formation due to
desmosome dissociation (pemphigus foliaceus (PF), pemphigus vulgaris (PV),
paraneoplastic pemphigus (PNP))
(b) Disruption of basement membrane adhesion – subepidermal blister formation
due to dermo-epidermal separation (bullous pemphigoid (BP), mucous membrane pemphigoid (MMP))
Desmosome Autoimmunity
emphigus Foliaceus (PF)
P
Pemphigus foliaceus is the most common autoimmune skin disease in cats that
accounts for about 1% of all skin diseases for which cats are seen by dermatologists [1]. Although the pathogenesis of feline PF has not been studied in
such extent as it has been in dogs, it is believed that, like in dogs and people,
antikeratinocyte IgG autoantibodies disrupt desmosomal adhesion between
keratinocytes and induce subcorneal blisters in a form of pustules (Fig. 1a).
Indeed, tissue-bound and circulating antikeratinocyte IgG have been detected in
the majority of cats with PF (Fig. 2d) [4, 5]. The major target autoantigen that
in people and dogs is desmoglein-1 and desmocollin-1, respectively, remains
unknown for feline PF.
Signalment
Cats of different breeds have been reported to suffer with PF, but a breed predisposition has not been confirmed yet. The most commonly reported breeds
Autoimmune Diseases
497
b
VetBooks.ir
a
c
e
d
f
Fig. 1 Feline pemphigus foliaceus – clinical lesions: (a) Pustule and scale-crust; (b) well-­
demarcated scale-crust suggesting a pustular origin on the concave pinna; (c) claw skin fold with
erythema, superficial erosions, scale-crust, and purulent exudate; (d) erosions and scale-crust on
the nasal planum and dorsal muzzle; (e) scale-crust around the areola; (f) erosions and scale-crust
on the footpad. (photo f – courtesy of Dr. Andrea Lamm)
498
P. Bizikova
b
c
d
PF cat: Serum anti-keratinocyte IgG
Healthy cat: Serum anti-keratinocyte IgG
VetBooks.ir
a
Fig. 2 Feline pemphigus foliaceus – histopathology and indirect immunofluorescence: (a)
Subcorneal pustule with acantholytic keratinocytes; (b) close-up of individualized and clustered
acantholytic keratinocytes (Courtesy of Dr. Keith Linder); (c) and (d) indirect immunofluorescence using healthy feline serum (c) and serum from a cat with pemphigus foliaceus (d). Note the
intercellular, web-like immunofluorescence pattern in the PF-affected cat sample (d) caused by
circulating anti-keratinocyte IgG antibodies
include domestic shorthaired, Siamese, Persian and Persian-crossbred, Burmese,
Himalayan, and domestic medium-haired cats [6]. Pemphigus foliaceus affects usually adult cats (median age of onset about 6 years), though the range varies greatly
(0.25–16 years) [4, 6–9]. A sex predilection has not been confirmed, but females
appear to be marginally over-represented according to a recent review [6]. In most
cats, a specific trigger precipitating the PF onset cannot be identified. Rare reports
of a drug-triggered PF and PF associated with a thymoma can be found in the literature [8, 10–16].
Clinical Signs
The primary skin lesion of feline PF is a subcorneal pustule, which, because of its
superficial nature, progresses rapidly into an erosion and crust. Indeed, the two latter skin lesions may represent the only clinical findings during the physical examination. Lesions are usually bilaterally symmetric with pinnae and claw skin folds
being the most commonly affected body areas (Fig. 1b and c) [6]. Claw skin folds
often exhibit accumulation of a thick purulent exudate, which is usually related to
secondary bacterial infections seen in this body region more often than in others
[17]. Other typically affected body areas include nasal planum, eyelids, pawpads,
VetBooks.ir
Autoimmune Diseases
499
and periareolar areas (Fig. 1 d, e). Typical pawpads lesions are scaling, crusting,
and hyperkeratosis, though they are usually not as prominent as in dogs (Fig. 1f).
Pustules, if found, can be seen at the periphery of the pawpads not in contact with
the ground. Most cats (81%) exhibit lesions on two or more body regions, while
lesions localized to a single body area are less common (19%). More than a half of
cats are pruritic and show systemic signs such as lethargy, fever, and/or anorexia.
Diagnostic Approach
The most critical and often challenging step in the diagnostic approach is the identification of a subcorneal pustular process. Indeed, while there are several erosive
skin diseases in cats, the list of diseases presenting with primary subcorneal pustules with acantholysis is limited to PF and to anecdotal reports of pustular dermatophytosis; the latter has been reported to exhibit minimal to no acantholysis
[18]. Bullous impetigo, a subcorneal pustular dermatitis with variable degree of
acantholysis caused by Staphylococcus aureus and pseudintermedius in people
and dogs, has not been described in cats yet [19, 20]. Lesions suggestive of a subcorneal pustular dermatitis include intact pustules, sharply demarcated, pinpoint
to few millimeters large; superficial erosions; or scaling and crusting (Fig. 1a, b).
The acantholytic nature of the disease can be confirmed by cytology taken from
an intact pustule, from the underneath of a crust with an active erosion and exudation, or from the caseous pus around nails and/or by a biopsy of similar lesions.
It is important to include crusts in the biopsy sample. Microscopic examination
of biopsy samples reveals acantholytic keratinocytes, usually numerous, within a
neutrophilic or mixed neutrophilic and eosinophilic, subcorneal or intragranular
pustule (Fig. 2a, b). Ghost acantholytic cells can be found within the crusts, and, in
many cases, may be the only histological evidence of the disease process. Aerobic
bacterial culture should be considered in cases in which infection cannot be clinically ruled out, and fungal culture and special stains should be considered in cases
in which dermatophytosis is suspected, particularly if a pustular folliculitis, lymphocytic mural folliculitis, and/or prominent hyperkeratosis is present in biopsy
samples. An immunological testing for antikeratinocyte autoantibodies by direct
or indirect immunofluorescence is not commercially available, nor the sensitivity
and, particularly, specificity of such tests is known. Therefore, the current diagnosis
of PF is based on the combination of (i) skin lesion character and distribution, (ii)
exclusion of an infection, and (iii) supportive cytology and/or histopathology confirming acantholytic pustular dermatitis [21].
Treatment
Cats with PF have usually positive response to treatment, and the majority of them
(93%) reach disease control (cessation of active lesions and healing of original
lesions) within a few weeks (median time, 3 weeks) [6]. In most cats, the disease control can be achieved by a glucocorticoid monotherapy (e.g., prednisolone, 2–4 mg/
kg/day; triamcinolone aceponate, 0.2–0.6 mg/kg/day; dexamethasone, 0.1–0.2 mg/
kg/day). The dosage reduction is recommended only once the disease has been inactive for at least 2 weeks and most original skin lesions have healed (20–25% dosage
VetBooks.ir
500
P. Bizikova
reduction every 2–4 weeks, though faster reduction is possible). The use of nonsteroidal agents is implemented in cats: (i) in which disease control is not achieved
within 4 weeks using appropriate glucocorticoid dosages, (ii) exhibiting severe
adverse effects related to glucocorticoids, or (iii) in which the dosage of glucocorticoids cannot be significantly reduced. Non-steroidal drugs reported to induce disease
control in cats include ciclosporin (5–10 mg/kg/day), chlorambucil (0.1–0.3 mg/kg/
day), azathioprine (1.1 mg/kg every other day) [7], and aurothioglucose (0.5 mg/
kg/week). The two latter are not used commonly either because of the high risk of
a bone marrow suppression (azathioprine) or its unavailability on the market (aurothioglucose). The risk of side effects in azathioprine-treated cats is dose-dependent,
and, anecdotally, lower dosages (e.g., 0.3 mg/kg every other day) have been reported
to be successful in managing other immune-mediated diseases [22].
According to the literature review, only a minority of cats (15%) appears to
achieve a long-term disease remission off drugs [6]. Most cats require a long-term
medical management with glucocorticoids or non-steroidal drugs such as ciclosporin or chlorambucil. The median maintenance dosages are usually lower than those
required for the induction of the disease control (e.g., prednisolone, 0.5 mg/kg/day;
dexamethasone, 0.03 mg/kg/day; or ciclosporin, 5 mg/kg/day). A combination of
doxycycline and niacinamide has also been reported to be an effective maintenance
therapy in cats with PF [17]. In refractory cases, other immunosuppressants (e.g.,
mycophenolate, leflunomide) could be considered, though the evidence of efficacy
of these drugs in feline PF is currently lacking.
Box 1: Feline PF Treatment Outline Using Similar Principles than Those Used in
Human Pemphigus [42]
(I) Induce rapid disease control (i.e., time at which new lesions cease to
form and old lesions start to heal)
First line treatment: prednisolone or methylprednisolone 2–4 mg/kg/
day (or its equivalent; e.g., triamcinolone aceponate, dexamethasone)
until disease control is reached.
(II) Start gradual glucocorticoid dosage reduction (25% every 2 weeks)
once the end of consolidation phase is reached (i.e., time at which no
new lesions have developed for at least 2 weeks and approximately 80%
of original lesions have healed). Consider also to taper to every-other-­
day administration before reducing the daily dose.
(III) Continue with the gradual glucocorticoid dosage reduction until the
lowest effective dosage is identified or the cat is able to remain in remission off of drugs (a long-term remission off drugs has been reported in
about 15% of cats).
III.a.Consider topical glucocorticoids (e.g., hydrocortisone aceponate)
or tacrolimus to control minor, localized flares.
III.b.In case of a more severe flare-up, increase the dose of glucocorticoids back to the second to the last effective dosage. If the flare-­up
VetBooks.ir
Autoimmune Diseases
501
cannot be controlled within 2 weeks, increase the glucocorticoid
dosage back to the initial immunosuppressive dosage.
(IV) Add non-steroidal immunosuppressive drug∗ if:
IV.a.The dosage of glucocorticoid cannot be reduced enough to limit
the risk of side effects associated with a long-term glucocorticoid
treatment.
IV.b.
The patient suffers with intolerable side effects caused by
glucocorticoids.
IV.c.Disease control cannot be achieved with glucocorticoid monotherapy within 4–6 weeks.
* Frequently selected non-steroidal immunosuppressive drugs in cats
with PF are ciclosporin (5–10 mg/kg/day) or chlorambucil (0.2 mg/kg/every
other day).
(V) V. Maintenance treatment:
V.a.Maintain the disease with the lowest possible dosage of drug(s);
attempt to reduce the dosage of glucocorticoids as much as
possible, or to complete replace them with a non-steroidal
immunosuppressant.
V.b.Monitor for side effects (usually complete blood count, chemistry panel, urinalysis, and urine culture every 6–12 months,
though the frequency and type of tests depends on the used
drug(s), age of the cat, and its general health status).
V.c.Avoid potential flare-up triggers (e.g., UV light, etc.).
Pemphigus Vulgaris (PV)
In contrast to people, PV is considered to check if this should autoimmune dermatoses in animals, including the cat [4]. Because of the small number of described
cases, breed, age, and sex predilections cannot be reliably estimated in cats. The
clinical and histological homology between feline, canine, and human PV suggests
similar pathomechanism; however, while desmoglein-3 has been confirmed to be
the major target autoantigen in human and canine PV, the major target antigen in
feline PV remains unknown.
Clinical Signs
Similarly to people, the primary lesion of animal PV is a flaccid vesicle rapidly
progressing to a deep erosion (Fig. 3a–c). Erosions are seen more often due to the
fragile nature of the vesicles, and further epithelial splitting beyond the preexisting erosion, even extending to a great distance (marginal Nikolsky’s signs), can be
elicited by pulling on the blister remnant. Crusts can develop over lesions at the
mucocutaneous junctions or haired skin. The current knowledge about the lesion
distribution in cats is extracted from less than a handful of described cases in the
literature and from anecdotal reports [4, 23]. Like in people and dogs, lesions frequently involve the oral cavity, especially gums and hard palate, lips, nasal planum,
P. Bizikova
VetBooks.ir
502
Fig. 3 Pemphigus vulgaris: Cats with pemphigus vulgaris with deep erosions affecting the (a) oral
cavity, lips, and nasal philtrum; (b) periocular region; and (c) pawpads. A histopathology of feline
pemphigus vulgaris showing the classic suprabasilar acantholysis (d). (Courtesy of Dr. Karen
Trainor)
and philtrum (Fig. 3a, b). Haired skin involvement, as described in people, dogs,
and horses [20, 24], and pawpad involvement (Fig. 3c) have been observed as well.
Sialorrhea, halitosis, dysphagia, lethargy, and enlarged submandibular lymph nodes
are common.
Diagnostic Approach
Because intact vesicles are rarely found, primary erosive diseases, especially those
affecting oral cavity and mucocutaneous junctions, present the major differential
diagnoses for feline PV. These include common diseases such as viral stomatitis
caused by herpesvirus or calicivirus and chronic ulcerative stomatitis or rare diseases such as autoimmune subepidermal blistering skin diseases. The diagnosis of
PV is confirmed by a biopsy, which shows suprabasilar acantholysis with basal cells
remaining attached to the basement membrane (Fig. 3d). Samples for histopathology should be collected from the margin of the blister or erosion and should include
VetBooks.ir
Autoimmune Diseases
503
affected as well as intact tissue adjacent to the blister or erosion. The diagnostic
value of direct and indirect immunofluorescence for antikeratinocyte autoantibodies
has not been addressed in cats, and, so far, only tissue-bound, but not circulating,
antikeratinocyte antibodies have been uncovered in cats with PV [4, 23].
Treatment
The information about the treatment and outcome of feline PV is very limited. Oral
glucocorticoids (4–6 mg/kg/day of prednisolone) were reported to induc
Download